EP3416661A1 - Association d'une immunothérapie et d'une thérapie de contrôle des cytokines pour le traitement du cancer - Google Patents

Association d'une immunothérapie et d'une thérapie de contrôle des cytokines pour le traitement du cancer

Info

Publication number
EP3416661A1
EP3416661A1 EP17752797.5A EP17752797A EP3416661A1 EP 3416661 A1 EP3416661 A1 EP 3416661A1 EP 17752797 A EP17752797 A EP 17752797A EP 3416661 A1 EP3416661 A1 EP 3416661A1
Authority
EP
European Patent Office
Prior art keywords
cells
another embodiment
cell
apoptotic
car
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP17752797.5A
Other languages
German (de)
English (en)
Other versions
EP3416661A4 (fr
Inventor
Shai Novik
Dror Mevorach
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Enlivex Therapeutics RDO Ltd
Original Assignee
Enlivex Therapeutics Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Enlivex Therapeutics Ltd filed Critical Enlivex Therapeutics Ltd
Publication of EP3416661A1 publication Critical patent/EP3416661A1/fr
Publication of EP3416661A4 publication Critical patent/EP3416661A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464403Receptors for growth factors
    • A61K39/464406Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464419Receptors for interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/59Reproductive system, e.g. uterus, ovaries, cervix or testes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/11Coculture with; Conditioned medium produced by blood or immune system cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • compositions and methods thereof for maintaining or increasing the proliferation rate of chimeric antigen receptor-expressing T-cells during CAR T-cell cancer therapy are disclosed herein. Further, disclosed herein are compositions and methods thereof for increasing the efficacy of chimeric antigen receptor T-cell cancer therapy, wherein the incidence of a subject experiencing cytokine release syndrome or a cytokine storm is reduced or inhibited.
  • Methods disclosed herein include those comprising administration of CAR T-cells and an additional agent comprising apoptotic cells, an apoptotic cell supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • CAR T-cells chimeric antigen receptor
  • ALL advanced acute lymphoblastic leukemia
  • lymphoma lymphoma
  • CRS cytokine release syndrome
  • cytokine storm in which the infused, activated T-cells produce a systemic inflammatory response in which there is a rapid and massive release of cytokines into the bloodstream, leading to dangerously low blood pressure, high fever and shivering.
  • cytokine storm a.k.a. cytokine cascade or hypercytokinemia
  • cytokine cascade or hypercytokinemia in which there is a positive feedback loop between cytokines and white blood cells with highly elevated levels of cytokines.
  • This can lead to potentially life-threatening complications including cardiac dysfunction, adult respiratory distress syndrome, neurologic toxicity, renal and/or hepatic failure, pulmonary edema and disseminated intravascular coagulation.
  • CAR-modified T cells with specificity against CD 19 have demonstrated dramatic promise against highly refractory hematologic malignancies.
  • Clinical responses with complete remission rates as high as 90% have been reported in children and adults with relapsed/refractory acute lymphoblastic leukemia (ALL).
  • ALL acute lymphoblastic leukemia
  • very significant toxicity has been observed and as many as 30% of subject administered CAR-T cells develop severe forms of CRS and possibly related neurotoxicity.
  • CRS is occurring due to large secretion of proinflammatory cytokines mainly from macrophages/monocytes, and resembles macrophage activating syndrome and hemophagocytosis, which is in response to CAR-T secreting interferon- gamma (IFN- ⁇ ) and possibly additional cytokines.
  • IFN- ⁇ interferon- gamma
  • corticosteroids biological therapies such as anti-IL6 therapies and anti- inflammatory drugs are being evaluated to control cytokine release syndrome in patients administered CAR T-cell therapy.
  • steroids may affect CAR T-cells' activity and/or proliferation and put the patients in danger of sepsis and opportunistic infections.
  • Anti-inflammatory drugs may not be effective in controlling cytokine release syndromes or cytokine storms, because the cytokine storm includes a very large number of cytokines while there is limited ability to infuse patients with anti-inflammatory drugs. Novel strategies are needed to control cytokine release syndromes, and especially cytokine storms, in order to realize the potential of CAR T-cell therapy.
  • Cytokine storms are also a problem after other infectious and non-infectious stimuli.
  • cytokines such as interleukin-1 (IL-1), IL-6, interferon- gamma (IFN- ⁇ ), and tumor necrosis factor-a (TNFa)
  • IFN- ⁇ interferon-gamma
  • TNFa tumor necrosis factor-a
  • IFN- ⁇ also excited macrophages, which in turn may secrete vast quantities of pro-inflammatory cytokines including IL-6 and TNF-a.
  • CRS is the most common potentially severe toxicity associated with CAR T cells, but it occurs with other therapies that engage T cells to kill cancer cells, including bispecific T-cell- engaging (BiTE) antibodies such as blinatumomab, and even in non-T cell therapies such as rituxan. Nevertheless, occurrence in 80-100% of patients is unique to CAR T cells, where 30% of patients with ALL have a severe form of toxicity that can be fatal in some patients.
  • BiTE bispecific T-cell- engaging
  • Neurotoxicity which could be regarded separately or as part of the syndrome, includes mental status changes, reversible delirium, and seizure-like activity. Patients may develop a gradual progression of confusion, word-finding difficulty, and aphasia, and ultimately become obtunded. In three cases, these neurologic complications required intubation and mechanical ventilation for airway protection. Patients with neurologic complications were evaluated with CT and MRI of the brain, which did not depict changes apart from possible leukoencephalopathy in some cases, as well as electroencephalograms (EEGs) and lumbar punctures. The EEGs confirmed seizure-like activity, which resolved after antiepileptic treatment.
  • CSF cerebrospinal fluid
  • HHLH hemophagocytic lymphohistiocytosis
  • MAS macrophage activating syndrome
  • Tocilizumab is an IL-6 receptor antagonist that is used to treat rheumatologic disorders. It was used to treat CRS-related toxicities in clinical trials, and is now widely used off-label for toxicity following CAR T-cell infusions. Tocilizumab may lessen or abrogate CRS-related toxicities following CAR T-cell infusions. Uncontrolled studies suggest that treating ALL patients, complete remissions still occur when they receive tocilizumab to treat CRS caused by CAR T cells.
  • tocilizumab might subtly impair the depth or duration of anti-malignancy responses caused by CAR T cells as formal studies of the impact of tocilizumab on anti-malignancy outcomes have not been performed.
  • most published experience with tocilizumab is with ALL.
  • Tocilizumab might impair the efficacy of CAR T cells against lymphoma or other malignancies even if it does not impair the activity of CAR T cells against ALL.
  • tocilizumab should not be administered for neurologic toxicity because of concerns about its ability to cross the blood brain barrier, and experience in an admittedly very small number of patients that tocilizumab did not ameliorate neurologic toxicity.
  • CAR T cells can cause additional, less significant toxicity by several mechanisms. If the tumor-associated antigen to which the CAR is targeted is expressed on normal tissues, those tissues may be damaged, as is the case with normal B cells being depleted by anti-CD19 CAR T cells. CAR T cells may damage normal tissues by unexpectedly cross-reacting with a protein that is not expressed on tumor cells. Acute anaphylaxis and tumor lysis syndrome (TLS) have occurred following infusion of CAR T cells; however, these toxicities are by far less frequent in comparison to CRS.
  • TLS tumor lysis syndrome
  • CAR T-cell efficacy may be dependent on a number of factors including persistence and survival of the genetically modified CAR T-cells, cell dose-as the final steady-state number of cells appears to be patient specific, and loss or down-regulation of expression of targeted antigens.
  • Novel strategies are therefore needed, which maintain or increase the efficacy of CAR T- cell therapies while at the same time controlling safety issues including cytokine release syndrome and especially cytokine storms. Further, there is a need to develop in vitro and in-vivo models of CRS with and without CAR-modified T cells. Disclosed herein are in vitro and in vivo models of CRS in which the effects of early apoptotic cell populations were tested for their effectiveness on cytokine release and CAR T-cell toxicity.
  • a method of maintaining or increasing the proliferation rate of chimeric antigen receptor-expressing T-cells (CAR T-cell) during CAR T-cell cancer therapy comprising the step of administering a composition comprising apoptotic cells or an apoptotic cell supernatant to a subject undergoing CAR T-cell therapy, and wherein said proliferation rate is maintained or increased in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant.
  • the method does not reduce or inhibit the efficacy of said CAR T-cell cancer therapy.
  • the incidence of cytokine release syndrome (CRS) or a cytokine storm in said subject is inhibited or reduced compared with a subject not administered said apoptotic cells or said apoptotic cell supernatant.
  • CRS cytokine release syndrome
  • said apoptotic cells comprise apoptotic cells in an early- apoptotic state.
  • said apoptotic cells are autologous to the subject being treated by said CAR T-cell therapy or are pooled third-party donor cells.
  • administration of said composition comprising said apoptotic cells or said apoptotic cell supernatant occurs prior to, concurrent with, or following the CAR T-cell therapy.
  • administration of said apoptotic cells or said apoptotic supernatant occurs prior to, concurrent with, or following the CAR T-cell therapy.
  • the apoptotic cell supernatant is an apoptotic cell-white blood cell supernatant, wherein white blood cells are co-cultured with the apoptotic cells prior to collection of the apoptotic cell- white blood cell supernatant.
  • the white blood cells are selected from the group consisting of phagocytes, macrophages, dendritic cells, monocytes, B cells,
  • T cells T cells, and NK cells.
  • the method maintains or increases the levels of IL-2 in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant.
  • a method of increasing the efficacy of chimeric antigen receptor T-cell (CAR T-cell) cancer therapy comprising the step of administering CAR T-cells and an additional agent selected from the group comprising apoptotic cells, an apoptotic cell supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, wherein said efficacy said CAR T-cells is increased in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said additional agent.
  • an additional agent selected from the group comprising apoptotic cells, an apoptotic cell supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof
  • the level of production of at least one pro-inflammatory cytokine is reduced compared with the level of said pro-inflammatory cytokine in a subject received CAR T-cell cancer therapy and not administered a composition comprising said agent.
  • the pro-inflammatory cytokine comprises IL-6.
  • apoptotic cells or an apoptotic cell supernatant when administered, said method maintains or increases the levels of IL-2 in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant.
  • the incidence of cytokine release syndrome (CRS) or a cytokine storm in said subject is inhibited or reduced compared with a subject not administered said additional agent.
  • CAR T-cells and said additional agent or any combination thereof are comprised in a single composition.
  • said CAR T-cell and said additional agent or any combination thereof are comprised in at least two compositions.
  • said additional agent or any combination of agents thereof is comprised in a composition not including said CAR T-cells, the administration of said composition comprising said agent or agents occurs prior to, concurrent with, or following administration of said CAR T-cells.
  • said apoptotic cells comprise apoptotic cells in an early-apoptotic state.
  • said apoptotic cells are autologous to a subject being treated by said CAR T- cell therapy or are pooled third-party donor cells.
  • the administration of said composition comprising said agent occurs prior to, concurrent with, or following administration of said CAR T-cells.
  • said apoptotic cell supernatant is an apoptotic cell- white blood cell supernatant, wherein white blood cells are co-cultured with the apoptotic cells prior to collection of the apoptotic cell-white blood cell supernatant.
  • the provided white blood cells are selected from the group consisting of phagocytes, macrophages, dendritic cells, monocytes, B cells, T cells, and NK cells.
  • a method of treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject comprising the step of administering chimeric antigen receptor-expressing T-cells (CAR T-cell) and an additional agent, said additional agent comprising apoptotic cells, apoptotic supernatants or a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, wherein said method treats, prevents, inhibits, reduces the incidence of, ameliorates or alleviates a cancer or a tumor in said subject compared with a subject administered CAR T-cells and not administered said additional agent.
  • CAR T-cell chimeric antigen receptor-expressing T-cells
  • said method has increased efficacy treating, preventing, inhibiting, reducing the incidence of, ameliorating or alleviating said cancer or said tumor in said subject compared with a subject administered CAR T-cells and not administered said additional agent.
  • the level of production of at least one pro-inflammatory cytokine is reduced compared with the level of said pro-inflammatory cytokine in a subject administered said CAR T-cells and not administered a composition comprising said agent.
  • said pro-inflammatory cytokine comprises IL-6.
  • said additional agent comprises apoptotic cells or an apoptotic cell supernatant, said method maintains or increases the levels of IL-2 in the subject compared with a subject administered said CAR T-cells and not administered said apoptotic cells or said apoptotic cell supernatant.
  • said CAR T-cells and said additional agent or any combination thereof are comprised in a single composition.
  • said CAR T-cells and said additional agent or any combination thereof are comprised in at least two compositions.
  • said additional agent or any combination of agents thereof is comprised in a composition not including said CAR T-cells, the administration of said composition comprising said agent or agents occurs prior to, concurrent with, or following administration of said CAR T-cells.
  • the administration of said additional agent occurs prior to, concurrent with, or following the administration of said CAR T-cells.
  • said apoptotic cells comprise apoptotic cells in an early-apoptotic state.
  • said apoptotic cells are autologous to said subject or are pooled third-party donor cells.
  • said apoptotic cell supernatant is an apoptotic cell- white blood cell supernatant, wherein white blood cells are co-cultured with the apoptotic cells prior to collection of the apoptotic cell-white blood cell supernatant.
  • the provided white blood cells are selected from the group consisting of phagocytes, macrophages, dendritic cells, monocytes, B cells, T cells, and NK cells.
  • Figure 1 Flow chart presenting the steps during one embodiment of a manufacturing process of an early apoptotic cell populations, wherein anti-coagulants were included in the process.
  • Figures 2A-2B Schematic showing standard CAR T-cell therapy (Figure 2A) and embodiments of a method of safe and efficacious CAR T-cell cancer therapy in a patient using patients' own cells (autologous) ( Figure 2B) to produce apoptotic cells or an apoptotic cell supernatant.
  • Figure 3 Schematic showing embodiment of a method of safe and efficacious CAR T-cell cancer therapy in a patient, using donor cells to produce apoptotic cells or an apoptotic supernatant.
  • Figure 4. Verification of Transduction of T cells showing flow cytometry results of anti- CD 124 analysis of transduced T4 + CAR-T cells.
  • Figure 5 SKOV3-luc growth in 24-well plate. 3.8xl0 4 -3.8xl0 5 SKOV3-luc cells/well were plated in 24-well plates and luciferase activity was recorded daily.
  • FIG. 7 Apoptotic Cells do not abrogate T4 + CAR-T cells anti-tumor activity. Results are based on a cytotoxicity assay, wherein a monolayer of SKOV3-luc cells were cultured either with non-transduced T cells or with T4 + CAR-T cells in the presence of a vehicle (Hartmann solution), or apoptotic cells (Apocell), or a supernatant of apoptotic cells (ApoSup), or supernatant of co-culture of apoptotic cells and monocytes/macrophages (ApoMon Sup).
  • a vehicle Hardtmann solution
  • Apocell apoptotic cells
  • ApoSup a supernatant of apoptotic cells
  • ApoMon Sup supernatant of co-culture of apoptotic cells and monocytes/macrophages
  • results shown here demonstrate the effect of co-culture of SKOV3-luc and human monocytes/macrophages were exposed to apoptotic cells (ApoCell), or ApoCell supernatant (ApoSup), or apoptotic cells and monocyte/macrophage co-culture (ApoMon Sup) in the presence of cancer and CAR- 19,
  • Figures 9A - 9J.Apoptotic cells prevent cytokine storm in in vitro model of cytokine storm induced in LPS-Sterile model of macrophage activation syndrome in a cancer environment.
  • Figure 9A shows the reduction of LPS induced IL-10 levels in the macrophage activation syndrome model in the presence of cancer following administration of Apocells at a macrophage/monocyte:Apocell ratio of 1 :8 and 1:16, at two time periods (6 hours and 24 hours).
  • Figure 9B shows the reduction of LPS induced IL-6 levels in the macrophage activation syndrome model following administration of Apocells in the presence of cancer and CAR-19, at a macrophage/monocyte:Apocell ratio of 1 :8 andl :16, at two time periods (6 hours and 24 hours).
  • Figure 9C shows the reduction of LPS induced ⁇ - ⁇ levels in the macrophage activation syndrome model in the presence of cancer and CAR-19, following administration of Apocells at a macrophage/monocyte:Apocell ratio of 1 :8 andl :16, at two time periods (6 hours and 24 hours).
  • Figure 9D shows the reduction of LPS induced IL-8 levels in the macrophage activation syndrome model in the presence of cancer and CAR-19, following administration of Apocells at a macrophage/monocyte:Apocell ratio of 1 :8 andl :16, at two time periods (6 hours and 24 hours).
  • Figure 9E shows the reduction of LPS induced TNF-a levels in the macrophage activation syndrome model in the presence of cancer and CAR-19, following administration of Apocells at a macrophage/monocyte: Apocell ratio of 1:8 andl:16, at TWO time periods (6 hours and 24 hours).
  • Figure 9F shows the reduction of LPS induced ⁇ -1 ⁇ levels in the macrophage activation syndrome model in the presence of cancer and CAR- 19, following administration of Apocells at a macrophage/monocyte:Apocell ratio of 1:4, 1 :8, 1 :16, 1 :32, and 1 :64 at 24 hours.
  • Figure 9G shows the reduction of LPS induced MCP-1 levels in the macrophage activation syndrome model in the presence of cancer and CAR- 19, following administration of Apocells at a macrophage/monocyte: Apocell ratio of 1 :4, 1:8, 1 :16, 1 :32, and 1 :64 at 24 hours.
  • Figure 9H shows the reduction of LPS induced IL-9 levels in the macrophage activation syndrome model in the presence of cancer and CAR- 19, following administration of Apocells at a macrophage/monocyte: Apocell ratio of 1 :8 andl:16, at two time periods (6 hours and 24 hours).
  • Figure 91 shows the increase of LPS induced IL-2R levels in the macrophage activation syndrome model in the presence of cancer and CAR-19, following administration of Apocells at a macrophage/monocyte: Apocell ratio of 1:4, 1 :8, 1:16, 1 :32, and 1 :64 at 24 hours.
  • Figure 9J shows that apoptotic cells do not down regulate IL-2 release from cells.
  • Empty bar (outline only) - 2.5 x 10 6 apoptotic cells per well; Black - 5 x 10 6 apoptotic cells per well; Grey - 10 xlO 6 apoptotic cells per well.
  • Figure 10 Effect of Apoptotic Cells or Apoptotic Cell Supernatant or a co-culture of Apoptotic cells and Monocytes following LPS exposure during CAR T-cell treatment mimicking CAR T-cell clinical therapy. Extremely high secretion of IL-6 was documented when lipopolysaccharides (LPS) were added to the cytotoxic assay. Results show that exposure to Apoptotic cells (Apocell), or supernatant of apoptotic cells (ApoSup) or supernatant of co-culture of apoptotic cells and monocytes/macrophages (ApoMon Sup), down regulated IL-6, wherein IL-6 was reduced to acceptable levels.
  • LPS lipopolysaccharides
  • Figures 11A-11B Weight and Tumor Size in Mice at time of Culling.
  • Figure 11A shows Weight change over the experimental time period. Blue-control no 4.5xl0 6 SKOV3-luc cells administrated. Red- 0.5x10 6 SKOV3-luc cells. Green-l .OxlO 6 SKOV3-luc cells. Purple-4.5xl0 6 SKOV3-luc cells
  • Figure 11B presents a representative SKOV3-luc tumor for a mouse receiving 4.5x10 6 SKOV3-luc cells, 39 days after injection.
  • FIG. 12 SKOV3-luc Tumor Growth. Mice bearing SKOV3-luc tumors imaged by Bioluminescent imaging (BLI) are presented showing the differences between control (PBS) and inoculation with 0.5xl0 6 , lxlO 6 , and 4.5xl0 6 SKOV3-luc cells.
  • FIGS 13A-13D SKOV3-luc Tumor Burden. Quantification of bioluminescence (BLI) of SKOV3-luc tumors in vivo (See Figure 12). A 600 photon count cut-off was implemented as instructed by the manufacturer.
  • Figure 13A mice inoculated with 0.5x106 SKOV3-luc.
  • Figure 13B mice inoculated with 1x106 SKOV3-luc.
  • Figure 13C mice inoculated with 4.5x106 SKOV3- luc.
  • Figure 13D Average SKOV3-luc tumor growth.
  • FIG. 14 Cytotoxic Calibration for Raji Burkett Lymphoma Cells. Raji cells were plated at various cell densities, with cell lysis occurring immediately prior to centrifugation. The results show Raji cell number (x-axis) vs. at absorbance at 492 nm (y-axis). All cell numbers exhibited significant readings relative to the unlysed counterpart.
  • Figure 15 Addition of early apoptotic cells does not affect CAR T-cell anti- tumor activity. E/T ratio shows the CD19+CAR T-cell to HeLa cell ratio. Survival is of CD19+ Tumor cells. Filled circle CD19+ Hela; Empty triangle CD19+ Hela + Naive T cells; Filled triangle CD19+ Hela + CAR T-CD19; Empty circle CD19+ Hela + CAR T-CD19 +ApoCells.
  • FIG. 16 Cytokine Analysis (GM-CSF) in Raji Burkett Lymphoma Cells in the Presence and Absence of Apoptotic cells.
  • the bar graph presents the concentration measurements of cytokine GM-CSF (pg/ml) found in culture supernatants of Raji cells incubated in the presence of monocytes and LPS, followed by addition of Naive T-cells (Raji + Naive T), CD19+ CAR T-cells (Raji + CAR T), CD19+ CAR T-cells and apoptotic cells (ApoCell) at a ratio of 1 :8 CAR T-cells:ApoCells (Raji + CAR T+ ApoCell 1 :8), CD19+ CAR T-cells and apoptotic cells (ApoCell) at a ratio of 1 :32 CAR T-cells :ApoCells (Raji + CAR T+ ApoCell 1 :32), and CD19+ C
  • FIG. 1 Cytokine Analysis (TNF-alpha) in Raji Burkett Lymphoma Cells in the Presence and Absence of Apoptotic cells.
  • the bar graph presents the concentration measurements of cytokine TNF-alpha (TNF-a) (pg/ml) found in culture supernatants of Raji cells incubated in the presence of monocytes and LPS, followed by addition of Naive T-cells (Raji + Naive T), CD19+ CAR T-cells (Raji + CAR T), CD19+ CAR T-cells and apoptotic cells (ApoCell) at a ratio of 1 :8 CAR T- cells :ApoCells (Raji + CAR T+ApoCell 1 :8), CD19+ CAR T-cells and apoptotic cells (ApoCell) at a ratio of 1 :32 CAR T-cells:ApoCells (Raji + CAR T+ ApoCell 1 :
  • Figures 18A and 18B presents the experimental scheme to analyze the influence of apoptotic cells on CAR T-cell therapy.
  • SCID mice were injected on day 1 with Raji cancer cells, followed on day 6 by administration of CAR T-CD19 cells (CAR T-cell therapy) and Apoptotic cells.
  • Figure 18B shows that CAR T-cell therapy was not negatively influenced by coadministration of ApoCells.
  • Survival Curve SCID mice were injected with CD19+ Raji cells with or without addition of early apoptotic cells.
  • Figures 19A, 19B, and 19C show increased release of pro-inflammatory cytokines from a tumor, in a solid tumor in vivo model.
  • Figure 19A shows slight increase of IL-6 released from a solid tumor present in the peritoneum of BALB/c and SCID mice, wherein the IL-6 release is significantly increased in the presence of HeLa CAR-CD- 19 CAR T-cells.
  • Figure 19B shows a slight increase of IP-10 released from a solid tumor present in the peritoneum of BALB/c and SCID mice, wherein the IP-10 release is significantly increased in the presence of HeLa CAR- CD- 19 CAR T-cells
  • Figure 19C shows that surprisingly even TNF-a release is increased by in the presence of HeLa CAR-CD- 19 CAR T-cells.
  • a method of maintaining or increasing the proliferation rate of chimeric antigen receptor-expressing T-cells (CAR T-cell) during CAR T-cell cancer therapy comprising the step of administering a composition comprising apoptotic cells or an apoptotic cell supernatant to said subject, and wherein said proliferation rate is maintained or increased in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant.
  • the method does not reduce or inhibit the efficacy of said CAR T- cell cancer therapy.
  • the incidence of cytokine release syndrome (CRS) or a cytokine storm in said subject is inhibited or reduced compared with a subject not administered said apoptotic cells or said apoptotic cell supernatant.
  • CRS cytokine release syndrome
  • CRS occurs spontaneously. In another embodiment, CRS occurs in response to LPS. In another embodiment, CRS occurs in response to IF - ⁇ .
  • a method of increasing the efficacy of chimeric antigen receptor T-cell (CAR T-cell) cancer therapy comprising the step of administering CAR T-cells and an additional agent selected from the group comprising apoptotic cells, an apoptotic cell supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, wherein said efficacy said CAR T-cells is increased in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said additional agent.
  • an additional agent selected from the group comprising apoptotic cells, an apoptotic cell supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof
  • the level of production of at least one pro-inflammatory cytokine is reduced compared with the level of said pro-inflammatory cytokine in a subject received CAR T-cell cancer therapy and not administered a composition comprising said agent.
  • the pro-inflammatory cytokine comprises IL-6.
  • apoptotic cells or an apoptotic cell supernatant when apoptotic cells or an apoptotic cell supernatant is administered, said method increases the levels of IL-2 in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant.
  • said method when apoptotic cells or an apoptotic cell supernatant is administered, said method maintains the levels of IL-2 in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant.
  • said method when apoptotic cells or an apoptotic cell supernatant is administered, said method maintains or increases the levels of IL-2 in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant.
  • the incidence of cytokine release syndrome (CRS) or a cytokine storm in said subject is inhibited or reduced compared with a subject not administered said additional agent.
  • CAR T-cells and said additional agent or any combination thereof are comprised in a single composition. In another related embodiment, said CAR T-cell and said additional agent or any combination thereof are comprised in at least two compositions.
  • a method of treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject comprising the step of administering chimeric antigen receptor-expressing T-cells (CAR T-cell) and an additional agent, said additional agent comprising apoptotic cells, apoptotic supernatants or a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, wherein said method treats, prevents, inhibits, reduces the incidence of, ameliorates or alleviates a cancer or a tumor in said subject compared with a subject administered CAR T-cells and not administered said additional agent.
  • CAR T-cell chimeric antigen receptor-expressing T-cells
  • said method has increased efficacy treating, preventing, inhibiting, reducing the incidence of, ameliorating or alleviating said cancer or said tumor in said subject compared with a subject administered CAR T-cells and not administered said additional agent.
  • the level of production of at least one pro-inflammatory cytokine is reduced compared with the level of said pro-inflammatory cytokine in a subject administered said CAR T-cells and not administered a composition comprising said agent.
  • said pro-inflammatory cytokine comprises IL-6.
  • said additional agent comprises apoptotic cells or an apoptotic cell supernatant
  • said method increases the levels of IL-2 in the subject compared with a subject administered said CAR T-cells and not administered said apoptotic cells or said apoptotic cell supernatant.
  • said CAR T-cells and said additional agent or any combination thereof are comprised in a single composition.
  • said CAR T-cells and said additional agent or any combination thereof are comprised in at least two compositions.
  • the administration of said additional agent occurs prior to, concurrent with, or following the administration of said CAR T-cells.
  • said apoptotic cells comprise apoptotic cells in an early- apoptotic state.
  • said apoptotic cells are autologous to said subject or are pooled third-party donor cells.
  • said apoptotic cell supernatant is obtained by a method comprising the steps of (a) providing apoptotic cells, (b) culturing the cells of step (a), and (c) separating the supernatant from the cells.
  • said apoptotic cell supernatant is an apoptotic cell-white blood cell supernatant and said method further comprises the steps of: (d) providing white blood cells, (e) optionally, washing the apoptotic cells and the white blood cells, (f) co-culturing the apoptotic cells and the white blood cells, wherein steps (d)-(f) are in place of step (b).
  • the provided white blood cells are selected from the group consisting of phagocytes, macrophages, dendritic cells, monocytes, B cells, T cells, and NK cells.
  • apoptotic supernatants comprise a supernatant produced by culturing apoptotic cells with macrophages, wherein the macrophage ingests the apoptotic cells and the supernatant produced from this co-culturing is used.
  • apoptotic supernatants comprise a supernatant produced by culturing apoptotic cells, wherein the supernatant is produced from materials secreted by the apoptotic cells.
  • Immune-cell based therapies include natural killer cells therapies, dendrite cell therapies, and T-cell immunotherapies including those utilizing naive T-cells, effector T-cells also known as T-helper cells, cytotoxic T-cells, and regulatory T-cells (Tregs).
  • compositions comprising genetically modified immune cells
  • the genetically modified immune cell is a T-cell.
  • a T-cell is a naive T-cell.
  • a T-cell is a naive CD4 + T-cell.
  • a T-cell is a naive T-cell.
  • a T-cell is a naive CD8 + T- cell.
  • the genetically modified immune cell is a natural killer (NK) cell.
  • the genetically modified immune cell is a dendritic cell.
  • the genetically modified T-cell is a cytotoxic T lymphocyte (CTL cell).
  • CTL cell cytotoxic T lymphocyte
  • the genetically modified T-cell is a regulatory T-cell (Treg). In another embodiment, the genetically modified T-cell is a chimeric antigen receptor (CAR) T-cell. In another embodiment, the genetically modified T-cell is a genetically modified T-cell receptor (TCR) cell.
  • Treg regulatory T-cell
  • CAR chimeric antigen receptor
  • TCR genetically modified T-cell receptor
  • compositions comprising genetically modified immune cells and an additional agent selected from the group comprising apoptotic cells, an apoptotic cell supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • compositions comprising genetically modified immune cells, apoptotic cells, and an additional agent selected from the group comprising a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • compositions comprising genetically modified immune cells, an apoptotic cell supernatant, and an additional agent selected from the group comprising a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • the immune cells are cytotoxic.
  • cytotoxic cells for genetic modification can be obtained from bone marrow of the subject (autologous) or a donor (allogeneic).
  • the cells are obtained from a stem cell.
  • cytotoxic cells can be derived from human pluripotent stem cells such as human embryonic stem cells or human induced pluripotent T-cells.
  • induced pluripotent stem cells IPCs
  • pluripotent T-cells can be obtained using a somatic cell from the subject to which genetically modified cytotoxic cells will be provided.
  • immune cells may be obtained from a subject or donor by harvesting cells by venipuncture, by apheresis methods, by white cell mobilization followed by apheresis or venipuncture, or by bone marrow aspiration.
  • immune cells for example T-cell
  • T-cell generation and maintenance is affected by cytokines in vivo.
  • cytokines that affect generation and maintenance to T-helper cells in vivo comprise IL-1, IL-2, IL-4, IL-6, IL-12, IL-21 , IL-23, IL-25, IL-33, and TGF .
  • Treg cells are generated from naive T-cells by cytokine induction in vivo.
  • TGF- ⁇ and/or IL-2 play a role in differentiating naive T-cell to become Treg cells.
  • the presence of a cytokine selected from the group comprising IL-1, IL-2, IL-4, IL-6, IL-12, IL-21, IL-23, IL-25, IL-33, and TGF maintains or increases the proliferation rate or both, of T-cells in vivo.
  • a cytokine IL-2 and/or TGF maintains or increases the proliferation rate or both, of T-cells in vivo.
  • the presence of a cytokine selected from the group comprising IL-1, IL-2, IL-4, IL-6, IL-12, IL-21, IL-23, IL-25, IL-33, and TGF maintains or increases the proliferation rate or both, of CAR T-cells in vivo.
  • a cytokine IL-2 and/or TGF maintains or increases the proliferation rate or both, of CAR T-cells in vivo.
  • the presence of a cytokine selected from the group comprising IL-1, IL-2, IL-4, IL-6, IL-12, IL-21, IL-23, IL-25, IL-33, and TGF maintains or increases the proliferation rate or both, of TCR T-cells in vivo.
  • a cytokine IL-2 and/or TGF maintains or increases the proliferation rate or both, of TCR T-cells in vivo.
  • the presence of a cytokine selected from the group comprising IL-1, IL-2, IL-4, IL-6, IL-12, IL-21, IL-23, IL-25, IL-33, and TGF maintains or increases the proliferation rate or both, of T-reg cells in vivo.
  • a cytokine IL-2 and/or TGF maintains or increases the proliferation rate or both, of T-reg cells in vivo.
  • T-cells having an altered expression or form of STAT5B encoded protein or BACH2 encoded protein are maintained for an extended time period or have an increased proliferation rate or both.
  • said altered expression increases expression STAT5B polypeptide.
  • said altered expression increases expression of BACH2 polypeptide.
  • T-cells having an altered expression of a STAT5B encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • T-cells having an altered expression of a BACH2 encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • T- cells having an altered form of a STAT5B encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • T-cells having an altered form of a BACH2 encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • T-cells having an altered expression of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 1 year. In another embodiment, T-cells having an altered expression of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 2 years. In another embodiment, T-cells having an altered expression of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 3 years. In another embodiment, T-cells having an altered expression of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 4 years.
  • T-cells having an altered expression of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 5 years. In another embodiment, T-cells having an altered expression of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 10 years. In another embodiment, T-cells having an altered expression of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 20 years.
  • T-cells having an altered expression of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 1 year. In another embodiment, T-cells having an altered expression of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 2 years. In another embodiment, T-cells having an altered expression of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 3 years. In another embodiment, T-cells having an altered expression of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 4 years. In another embodiment, T-cells having an altered expression of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 5 years.
  • T-cells having an altered expression of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 10 years. In another embodiment, T-cells having an altered expression of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 20 years.
  • T-cells having an altered form of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 1 year. In another embodiment, T-cells having an altered form of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 2 years. In another embodiment, T-cells having an altered form of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 3 years. In another embodiment, T-cells having an altered form of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 4 years.
  • T-cells having an altered form of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 5 years. In another embodiment, T-cells having an altered form of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 10 years. In another embodiment, T-cells having an altered form of a STAT5B encoded protein maintain or increase their proliferation rate in vivo for greater than 20 years.
  • T-cells having an altered form of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 1 year. In another embodiment, T-cells having an altered form of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 2 years. In another embodiment, T-cells having an altered form of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 3 years. In another embodiment, T-cells having an altered form of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 4 years. In another embodiment, T-cells having an altered form of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 5 years.
  • T-cells having an altered form of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 10 years. In another embodiment, T-cells having an altered form of a BACH2 encoded protein maintain or increase their proliferation rate in vivo for greater than 20 years.
  • CAR T-cells having an altered expression of a STAT5B encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • CAR T-cells having an altered expression of a BACH2 encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • CAR T-cells having an altered form of a STAT5B encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • CAR T-cells having an altered form of a BACH2 encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo
  • TCR T-cells having an altered expression of a STAT5B encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • TCR T-cells having an altered expression of a BACH2 encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • TCR T-cells having an altered form of a STAT5B encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • TCR T-cells having an altered form of a BACH2 encoded protein are maintained for an extended time period or have an increased proliferation rate in vivo.
  • Treg-cells having an altered expression of a STAT5B encoded protein maintain or increase their proliferation rate in vivo.
  • Treg-cells having an altered expression of a BACH2 encoded protein maintain or increase their proliferation rate in vivo.
  • Treg-cells having an altered form of a STAT5B encoded protein maintain or increase their proliferation rate in vivo.
  • Treg-cells having an altered form of a BACH2 encoded protein maintain or increase their proliferation rate in vivo.
  • methods for maintaining or increasing the proliferation rate of a genetically modified immune cell comprising the step of administering apoptotic cells or an apoptotic supernatant.
  • methods for increasing the efficacy of a genetically modified immune cell are disclosed herein, wherein the method comprises the step of administering an additional agent comprising apoptotic cells, an apoptotic supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • methods for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor disclosed herein administer a genetically modified immune cell and an additional agent, wherein said additional agent comprises apoptotic cells, an apoptotic supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • chimeric antigen receptors are a type of antigen-targeted receptor composed of intracellular T-cell signaling domains fused to extracellular tumor-binding moieties, most commonly single -chain variable fragments (scFvs) from monoclonal antibodies.
  • CARs directly recognize cell surface antigens, independent of MHC-mediated presentation, permitting the use of a single receptor construct specific for any given antigen in all patients.
  • Initial CARs fused antigen-recognition domains to the CD3 ⁇ activation chain of the T-cell receptor (TCR) complex. While these first generation CARs induced T-cell effector function in vitro, they were largely limited by poor antitumor efficacy in vivo.
  • CAR iterations have included secondary costimulatory signals in tandem with CD3 ⁇ including intracellular domains from CD28 or a variety of TNF receptor family molecules such as 4-1BB (CD137) and OX40 (CD134) .
  • third generation receptors include two costimulatory signals in addition to CD3 ⁇ , most commonly from CD28 and 4-1BB.
  • Second and third generation CARs dramatically improved antitumor efficacy, in some cases inducing complete remissions in patients with advanced cancer.
  • a CAR T-cell is an immunoresponsive cell comprising an antigen receptor, which is activated when its receptor binds to its antigen.
  • the CAR T-cells used in the compositions and methods as disclosed herein are first generation CAR T-cells.
  • the CAR T-cells used in the compositions and methods as disclosed herein are second generation CAR T-cells.
  • the CAR T-cells used in the compositions and methods as disclosed herein are third generation CAR T-cells.
  • the CAR T-cells used in the compositions and methods as disclosed herein are fourth generation CAR T-cells.
  • each generation of CAR T-cells is more potent than the CAR T-cells of earlier generations.
  • first-generation CARs have one signaling domain, typically the cytoplasmic signaling domain of the CD3 TC33 ⁇ 4 chain.
  • the CAR T-cells as disclosed herein are second generation CAR T- cells.
  • CAR T-cells as disclosed herein comprise a tripartite chimeric receptor (TPCR).
  • TPCR tripartite chimeric receptor
  • CAR T-cells as disclosed herein comprise one or more signaling moieties that activate naive T-cells in a co-stimulation independent manner.
  • the CAR T-cells further encode one or more members of the tumor necrosis factor receptor family, which in one embodiment, is CD27, 4-1BB (CD137), or OX40 (CD134), or a combination thereof.
  • Third-generation CAR T-cells attempt to harness the signaling potential of 2 costimulatory domains: in one embodiment, the CD28 domain followed by either the 4- IBB or OX-40 signaling domains.
  • the CAR T-cells used in the compositions and methods as disclosed herein further encode a co-stimulatory signaling domain, which in one embodiment is CD28.
  • the signaling domain is the CD3 ⁇ -chain, CD97, GDI la-CD18, CD2, ICOS, CD27, CD154, CDS, OX40, 4-1BB, CD28 signaling domain, or combinations thereof.
  • telomere length and replicative capacity correlate with the engraftment efficiency and antitumor efficacy of adoptively transferred T-cell lines.
  • CD28 stimulation maintains telomere length in T-cells.
  • CAR-modified T-cell potency may be further enhanced through the introduction of additional genes, including those encoding proliferative cytokines (ie, IL-12) or costimulatory ligands (ie, 4-1BBL), thus producing "armored” fourth-generation CAR-modified T- cells.
  • proliferative cytokines ie, IL-12
  • costimulatory ligands ie, 4-1BBL
  • “armored CAR T-cells” are CAR T-cells which are protected from the inhibitory tumor microenvironment.
  • the "armored” CAR technology incorporates the local secretion of soluble signaling proteins to amplify the immune response within the tumor microenvironment with the goal of minimizing systemic side effects.
  • the signaling protein signal is IL-12, which can stimulate T-cell activation and recruitment.
  • "armored" CAR technology is especially useful in solid tumor indications, in which microenvironment and potent immunosuppressive mechanisms have the potential to make the establishment of a robust anti-tumor response more challenging.
  • CAR T-cells are genetically modified to encode molecules involved in the prevention of apoptosis, the remodeling of the tumor microenvironment, induction of homeostatic proliferation, and chemokine receptors that promote directed T-cell homing.
  • CAR T-cell therapy used in the compositions and methods as disclosed herein is enhanced using the expression of cytokine transgenes, combination therapy with small molecule inhibitors, or monoclonal antibodies.
  • other strategies aimed at improving CAR T-cell therapy including using dual CARs and chemokine receptors to more specifically target tumor cells are to be considered part of the CAR T-cells and CAR T-cell therapy as disclosed herein.
  • the CAR T-cells of the compositions and methods as disclosed herein comprise a second binding domain that can lead to either an inhibitory or amplifying signal, in order to increase specificity of CAR T-cells for cancer cells versus normal cells.
  • a CAR T- cell can be engineered such that it would be triggered in the presence of one target protein, but if a second protein is present it would be inhibited.
  • it could also be engineered such that two target proteins would be required for maximal activation.
  • the CAR T-cells used in the compositions and methods as disclosed herein encode antibody-based external receptor structures and cytosolic domains that encode signal transduction modules composed of the immunoreceptor tyrosine-based activation motif.
  • the CAR T-cell further encodes a single-chain variable fragment (scFv) that binds a polypeptide that has immunosuppressive activity.
  • scFv single-chain variable fragment
  • the polypeptide that has immunosuppressive activity is CD47, PD-1, CTLA-4, or a combination thereof.
  • the CAR T-cell further encodes a single-chain variable fragment (scFv) that binds a polypeptide that has immunostimulatory activity.
  • the polypeptide that has immunostimulatory activity is CD28, OX-40, 4-1 BB or a combination thereof.
  • the CAR T-cell further encodes a CD40 ligand (CD40L), which, in one embodiment, enhances the immunostimulatory activity of the antigen.
  • a method as disclosed herein comprises obtaining immune cells from a subject, and genetically modifying the immune cells to express a chimeric antigen receptor.
  • a method as disclosed herein comprises obtaining immune cells from a subject, genetically modifying the immune cells to express a chimeric antigen receptor and combining with apoptotic cell population resulting in reduced cytokine production in a subject but substantially unaffected cytotoxicity relative to immune cells expressing a CAR not administered with an apoptotic cell population ( Figures 2A-2B and 3).
  • a method as disclosed herein comprises obtaining immune cells from a subject, genetically modifying the immune cells to express a chimeric antigen receptor and combining with an apoptotic cell supernatant or a composition comprising the supernatant, resulting in reduced cytokine production in a subject but substantially unaffected cytotoxicity relative to immune cells expressing a CAR not administered with an apoptotic cell supernatant.
  • administration of an apoptotic cell population or a supernatant from apoptotic cells does not reduce the efficacy of the immune cells expressing the chimeric antigen receptor.
  • one embodiment as disclosed herein relates to cytotoxic immune cells (e.g., NK cells or T-cells) comprising chimeric antigen receptors (CARs) whereby the cells retain their cytotoxic function.
  • CARs chimeric antigen receptors
  • the chimeric antigen receptor is exogenous to the T-cell.
  • the CAR is recombinantly expressed.
  • the CAR is expressed from a vector.
  • the T-cell utilized to generate CAR T-cells is a naive CD4 + T-cell. In another embodiment, the T-cell utilized to generate CAR T-cells is a naive CD8 + T-cell. In another embodiment, the T-cell utilized to generate CAR T-cells is an effector T-cell. In another embodiment, the T-cell utilized to generate CAR T-cells is a regulatory T-cell (Treg). In another embodiment, the T-cell utilized to generate CAR T-cells is a cytotoxic T-cell.
  • CAR T-cells have been described extensively in the literature, see for example Themelli et al. (2015) New Cell Sources for T Cell Engineering and Adoptive Immunotherapy. Cell Stem Cell 16: 357-366; Sharpe and Mount (2015) Genetically modified T cells in cancer therapy: opportunities and challenges.Diseas Models & Mechanisms 8:337-350; Han et al. (2013) Journal of Hematology & Oncology 6:47-53; Wilkie et al. (2010) J Bio Chem 285(33):25538-25544; and van der Stegen et al. (2013) J. Immunol 191 : 4589-4598.
  • CAR T-cells are available to order from a commercial source such as Creative Biolabs (NY USA), which provides custom construction and production services for Chimeric Antigen Receptors (CAR) and also provides premade CAR constructs stock, which can induce protective immunity encode by recombinant adenovirus vaccine.
  • a commercial source such as Creative Biolabs (NY USA)
  • CAR Chimeric Antigen Receptors
  • premade CAR constructs stock which can induce protective immunity encode by recombinant adenovirus vaccine.
  • T-cell receptors (TCRs) cells [0103] T-cell receptors (TCRs) cells
  • compositions and methods as disclosed herein utilize a designer T-cell receptor (TCR) cells in addition to or in place of CAR T-cells.
  • TCR is a multi-subunit transmembrane complex that mediates the antigen- specific activation of T-cells.
  • the TCR is composed of two different polypeptide chains.
  • the TCR confers antigenic specificity on the T cell, by recognizing an antigen epitope on the target cell, for example a tumor or cancer cell. Following contact with the antigen present on the tumor or cancer cell, T-cells proliferate and acquire the phenotype and function to allow them eliminate the cancer or tumor cells.
  • TCR T-cell therapy comprises introducing a T-cell receptor (TCR) that is specific to an epitope of a protein of interest into a T-cell.
  • the protein of interest is a tumor-associated antigen.
  • the genetically engineered TCR recognizes a tumor antigen epitope presented by the major histocompatibility complex (MHC) on the tumor cell along with T-cell activating domains.
  • MHC major histocompatibility complex
  • the T-cell receptors recognize antigens irrespectively of their intracellular or membrane localization.
  • TCRs recognize tumor cells that intracellularly express a tumor associated antigen.
  • TCRs recognize internal antigens.
  • TCRs recognize angiogenic factors.
  • an angiogenic factor is a molecule involved in the formation of new blood vessels.
  • Various genetically modified T-cell receptors and methods of their production are known in the art.
  • TCR T-cell therapy is used to treat, prevent, inhibit, ameliorate, reduce the incidence of, or alleviate a cancer or a tumor.
  • TCR T-cell therapy is used to treat, prevent, inhibit, ameliorate, reduce the incidence of, or alleviate advanced metastatic disease, including those with hematological (lymphoma and leukemia) and solid tumors (refractory melanoma, sarcoma).
  • the TCR T-cell therapy used in the compositions and methods as disclosed herein treat a malignancy listed in Table 1 of Sadelain et al., (Cancer Disco v. 2013 Apr; 3(4): 388-398).
  • the T-cell receptor is genetically modified to bind NY-ESO-1 epitopes, and the TCR-engineered T-cell is anti-NY-ESO-1.
  • the T-cell receptor is genetically modified to bind HPV-16 E6 epitopes, and the TCR-engineered T-cell is anti-HPV-16 E6.
  • the T-cell receptor is genetically modified to bind HPV- 16 E7 epitopes, and the TCR-engineered T-cell is anti-HPV-16 E7.
  • the T- cell receptor is genetically modified to bind MAGE A3/A6 epitopes, and the TCR-engineered T-cell is anti-MAGE A3/A6.
  • the T-cell receptor is genetically modified to bind MAGE A3 epitopes, and the TCR-engineered T-cell is anti-MAGE A3.
  • the T-cell receptor is genetically modified to bind SSX2 epitopes, and the TCR-engineered T-cell is anti-SSX2.
  • the T-cell receptor is genetically modified to bind a target antigen disclosed herein. Using the tools well known in the art, a skilled would appreciate that the T-cell receptor may be genetically modified to bind a target antigen present on a cancer or tumor cell, wherein the TCR-engineer T-cell comprises an anti-tumor or anti-cancer cell.
  • a method as disclosed herein comprises obtaining immune cells from a subject, and genetically modifying the immune cells to express a recombinant T-cell receptor (TCR).
  • a method as disclosed herein comprises obtaining immune cells from a subject, genetically modifying the immune cells to express a recombinant TCR and combining with an additional agent, wherein said additional agent comprises an apoptotic cell population, an apoptotic cell supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • the T-cell utilized to generate TCR T-cells is a naive CD4 + T-cell. In another embodiment, the T-cell utilized to generate TCR T-cells is a naive CD8 + T-cell. In another embodiment, the T-cell utilized to generate TCR T-cells is an effector T-cell. In another embodiment, the T-cell utilized to generate TCR T-cells is a regulatory T-cell (Treg). In another embodiment, the T-cell utilized to generate TCR T-cells is a cytotoxic T-cell.
  • TCR T-cells have been described extensively in the literature, see for example Sharpe and Mount (2015) ibid.; Essand M, Loskog ASI (2013) Genetically engineered T cells for the treatment of cancer (Review). J Intern Med 273: 166-181; and Kershaw et al. (2014) Clinical application of genetically modified T cells in cancer therapy. Clinical & Translational Immunology 3:1-7.
  • the CAR binds to an epitope of an antigen via an antibody or an antibody fragment that is directed to the antigen.
  • the antibody is a monoclonal antibody.
  • the antibody is a polyclonal antibody.
  • the antibody fragment is a single-chain variable fragment (scFv).
  • the TCR binds to an epitope of an antigen via a genetically modified T- cell receptor.
  • the CAR T-cells of the compositions as disclosed herein bind to a tumor associated antigen (TAA).
  • said tumor associated antigen is: Mucin 1, cell surface associated (MUCl) or polymorphic epithelial mucin (PEM), Arginine-rich, mutated in early stage tumors (Armet), Heat Shock Protein 60 (HSP60), calnexin (CANX), methylenetetrahydrofolate dehydrogenase (NADP+ dependent) 2, methenyltetrahydrofolate cyclohydrolase (MTHFD2), fibroblast activation protein (FAP), matrix metallopeptidase (MMP6), B Melanoma Antigen- 1 (BAGE-1), aberrant transcript of N-acetyl glucosaminyl transferase V (GnTV), Q5H943, Carcinoembryonic antigen (CEA), Pmel, Kallikrein-4, Mammaglobin
  • MUCl cell surface associated
  • the CAR binds to CD 19 or CD20 to target B cells in the case where one would like to destroy B cells as in leukemia.
  • CD19 is a B cell lineage specific surface receptor whose broad expression, from pro-B cells to early plasma cells, makes it an attractive target for the immunotherapy of B cell malignancies.
  • the CAR binds to RORl , CD22, or GD2.
  • the CAR binds to NY-ESO-1.
  • the CAR binds to MAGE family proteins.
  • the CAR binds to mesothelin.
  • the CAR binds to c-erbB2.
  • the CAR binds to mutational antigens that are tumor specific, such as BRAFV600E mutations and BCR-ABL translocations.
  • the CAR binds to viral antigens which are tumor-specific, such as EBV in HD, HPV in cervical cancer, and polyomavirus in Merkel cancer.
  • the CAR T-cell binds to Her2/neu.
  • the CAR T-cell binds to EGFRvIII.
  • the chimeric antigen receptor (CAR) T-cell binds the CD 19 antigen.
  • the CAR binds the CD22 antigen.
  • the CAR binds to alpha folate receptor.
  • the CAR binds to CAIX.
  • the CAR binds to CD20.
  • the CAR binds to CD23.
  • the CAR binds to CD24.
  • the CAR binds to CD30.
  • the CAR binds to CD33.
  • the CAR binds to CD38.
  • the CAR binds to CD44v6.
  • the CAR binds to CD44v7/8. In another embodiment, the CAR binds to CD 123. In another embodiment, the CAR binds to CD171. In another embodiment, the CAR binds to carcinoembryonic antigen (CEA). In another embodiment, the CAR binds to EGFRvIII. In another embodiment, the CAR binds to EGP-2. In another embodiment, the CAR binds to EGP-40. In another embodiment, the CAR binds to EphA2. In another embodiment, the CAR binds to Erb-B2. In another embodiment, the CAR binds to Erb-B 2,3,4. In another embodiment, the CAR binds to Erb-B3/4.
  • the CAR binds to FBP. In another embodiment, the CAR binds to fetal acetylcholine receptor. In another embodiment, the CAR binds to GD 2 - In another embodiment, the CAR binds to GD 3 . In another embodiment, the CAR binds to HER2. In another embodiment, the CAR binds to HMW-MAA. In another embodiment, the CAR binds to IL-l lRalpha. In another embodiment, the CAR binds toIL- 13Ralphal . In another embodiment, the CAR binds to KDR. In another embodiment, the CAR binds to kappa-light chain. In another embodiment, the CAR binds to Lewis Y.
  • the CAR binds to LI -cell adhesion molecule. In another embodiment, the CAR binds to MAGE-A1. In another embodiment, the CAR binds to mesothelin. In another embodiment, the CAR binds to CMV infected cells. In another embodiment, the CAR binds to MUC1. In another embodiment, the CAR binds to MUC16. In another embodiment, the CAR binds to NKG2D ligands. In another embodiment, the CAR binds to NY-ESO-1 (amino acids 157-165). In another embodiment, the CAR binds to oncofetal antigen (h5T4). In another embodiment, the CAR binds to PSCA.
  • the CAR binds to PSMA. In another embodiment, the CAR binds to ROR1. In another embodiment, the CAR binds to TAG-72. In another embodiment, the CAR binds to VEGF-R2 or other VEGF receptors. In another embodiment, the CAR binds to B7-H6. In another embodiment, the CAR binds to CA9. In another embodiment, the CAR binds to ⁇ ⁇ ⁇ integrin. In another embodiment, the CAR binds to 8H9. In another embodiment, the CAR binds to NCAM. In another embodiment, the CAR binds to fetal acetylcholine receptor.
  • the chimeric antigen receptor (CAR) T-cell targets the CD 19 antigen, and has a therapeutic effect on subjects with B-cell malignancies, ALL, Follicular lymphoma, CLL, and Lymphoma.
  • the CAR T-cell targets the CD22 antigen, and has a therapeutic effect on subjects with B-cell malignancies.
  • the CAR T-cell targets alpha folate receptor or folate receptor alpha, and has a therapeutic effect on subjects with ovarian cancer or epithelial cancer.
  • the CAR T-cell targets CAIX or G250/CAIX, and has a therapeutic effect on subjects with renal cell carcinoma.
  • the CAR T-cell targets CD20, and has a therapeutic effect on subjects with Lymphomas, B-cell malignancies, B-cell lymphomas, Mantle cell lymphoma and, indolent B-cell lymphomas.
  • the CAR T-cell targets CD23, and has a therapeutic effect on subjects with CLL.
  • the CAR T-cell targets CD24, and has a therapeutic effect on subjects with pancreatic adenocarcinoma.
  • the CAR T-cell targets CD30, and has a therapeutic effect on subjects with Lymphomas or Hodgkin lymphoma.
  • the CAR T-cell targets CD33, and has a therapeutic effect on subjects with AML.
  • the CAR T-cell targets CD38, and has a therapeutic effect on subjects with Non-Hodgkin lymphoma.
  • the CAR T-cell targets CD44v6, and has a therapeutic effect on subjects with several malignancies.
  • the CAR T-cell targets CD44v7/8, and has a therapeutic effect on subjects with cervical carcinoma.
  • the CAR T-cell targets CD 123, and has a therapeutic effect on subjects with myeloid malignancies.
  • the CAR T-cell targets CEA, and has a therapeutic effect on subjects with colorectal cancer.
  • the CAR T-cell targets EGFRvIII, and has a therapeutic effect on subjects with Glioblastoma.
  • the CAR T-cell targets EGP-2, and has a therapeutic effect on subjects with multiple malignancies.
  • the CAR T-cell targets EGP-40, and has a therapeutic effect on subjects with colorectal cancer.
  • the CAR T-cell targets EphA2, and has a therapeutic effect on subjects with Glioblastoma.
  • the CAR T-cell targets Erb-B2 or ErbB3/4, and has a therapeutic effect on subjects with Breast cancer and others, prostate cancer, colon cancer, various tumors.
  • the CAR T-cell targets Erb-B 2,3,4, and has a therapeutic effect on subjects with Breast cancer and others.
  • the CAR T-cell targets FBP, and has a therapeutic effect on subjects with Ovarian cancer.
  • the CAR T-cell targets fetal acetylcholine receptor, and has a therapeutic effect on subjects with Rhabdomyosarcoma.
  • the CAR T-cell targets GD 2 , and has a therapeutic effect on subjects with Neuroblastoma, melanoma, or Ewing's sarcoma.
  • the CAR T-cell targets GD 3 , and has a therapeutic effect on subjects with Melanoma.
  • the CAR T-cell targets HER2, and has a therapeutic effect on subjects with medulloblastoma, pancreatic adenocarcinoma, Glioblastoma, Osteosarcoma, or Ovarian cancer.
  • the CAR T-cell targets HMW-MAA, and has a therapeutic effect on subjects with Melanoma.
  • the CAR T-cell targets IL-1 IRalpha, and has a therapeutic effect on subjects with Osteosarcoma.
  • the CAR T-cell targets IL- 13Ralphal , and has a therapeutic effect on subjects with Glioma, Glioblastoma, or medulloblastoma.
  • the CAR T-cell targets IL-13 receptor alpha2, and has a therapeutic effect on subjects with several malignancies.
  • the CAR T-cell targets KDR, and has a therapeutic effect on subjects with tumors by targeting tumor neovasculature.
  • the CAR T-cell targets kappa-light chain, and has a therapeutic effect on subjects with B-cell malignancies (B-NHL, CLL).
  • the CAR T-cell targets Lewis Y, and has a therapeutic effect on subjects with various carcinomas or epithelial-derived tumors.
  • the CAR T-cell targets LI -cell adhesion molecule, and has a therapeutic effect on subjects with Neuroblastoma.
  • the CAR T-cell targets MAGE- A 1 or HLA-A1 MAGE Al, and has a therapeutic effect on subjects with Melanoma.
  • the CAR T-cell targets mesothelin, and has a therapeutic effect on subjects with Mesothelioma.
  • the CAR T-cell targets CMV infected cells, and has a therapeutic effect on subjects with CMV.
  • the CAR T-cell targets MUC1, and has a therapeutic effect on subjects with breast or ovarian cancer.
  • the CAR T-cell targets MUC16, and has a therapeutic effect on subjects with ovarian cancer.
  • the CAR T-cell targets NKG2D ligands, and has a therapeutic effect on subjects with myeloma, ovarian, and other tumors.
  • the CAR T-cell targets NY-ESO-1 (157-165) or HLA-A2 NY-ESO-1, and has a therapeutic effect on subjects with multiple myeloma.
  • the CAR T-cell targets oncofetal antigen (h5T4), and has a therapeutic effect on subjects with various tumors.
  • the CAR T-cell targets PSCA, and has a therapeutic effect on subjects with prostate carcinoma.
  • the CAR T-cell targets PSMA, and has a therapeutic effect on subjects with prostate cancer/tumor vasculature.
  • the CAR T-cell targets RORl , and has a therapeutic effect on subjects with B-CLL and mantle cell lymphoma.
  • the CAR T-cell targets TAG-72, and has a therapeutic effect on subjects with adenocarcinomas or gastrointestinal cancers.
  • the CAR T-cell targets VEGF-R2 or other VEGF receptors, and has a therapeutic effect on subjects with tumors by targeting tumor neo vasculature.
  • the CAR T-cell targets CA9, and has a therapeutic effect on subjects with renal cell carcinoma.
  • the CAR T-cell targets CD171 , and has a therapeutic effect on subjects with renal neuroblastoma.
  • the CAR T-cell targets NCAM, and has a therapeutic effect on subjects with neuroblastoma.
  • the CAR T-cell targets fetal acetylcholine receptor, and has a therapeutic effect on subjects with rhabdomyosarcoma.
  • the CAR binds to one of the target antigens listed in Table 1 of Sadelain et al. (Cancer Discov. 2013 Apr; 3(4): 388-398), which is incorporated by reference herein in its entirety.
  • CAR T-cells bind to carbohydrate or glycolipid structures.
  • the CAR binds to an angiogenic factor, thereby targeting tumor vasculature.
  • the angiogenic factor is VEGFR2.
  • the angiogenic factor is endoglin.
  • an angiogenic factor of the present invention is Angiogenin; Angiopoietin-1; Del-1 ; Fibroblast growth factors: acidic (aFGF) and basic (bFGF); Follistatin; Granulocyte colony-stimulating factor (G-CSF); Hepatocyte growth factor (HGF) /scatter factor (SF); Interleukin-8 (IL-8); Leptin; Midkine; Placental growth factor; Platelet-derived endothelial cell growth factor (PD-ECGF); Platelet-derived growth factor-BB (PDGF-BB); Pleiotrophin (PTN); Progranulin; Proliferin; Transforming growth factor-alpha (TGF-alpha); Transforming growth factor-beta (TGF-
  • an angiogenic factor is an angiogenic protein.
  • a growth factor is an angiogenic protein.
  • an angiogenic protein for use in the compositions and methods of the present invention is Fibroblast growth factors (FGF); VEGF; VEGFR and Neuropilin 1 (NRP-1); Angiopoietin 1 (Angl) and Tie2; Platelet-derived growth factor (PDGF; BB-homodimer) and PDGFR; Transforming growth factor-beta (TGF- ⁇ ), endoglin and TGF- ⁇ receptors; monocyte chemotactic protein-1 (MCP-1); Integrins ⁇ 3, ⁇ 5 and ⁇ 5 ⁇ 1 ; VE-cadherin and CD31 ; ephrin; plasminogen activators; plasminogen activator inhibitor- 1; Nitric oxide synthase (NOS) and COX- 2; AC133; or Idl/Id3.
  • FGF Fibroblast growth factors
  • VEGF
  • an angiogenic protein for use in the compositions and methods of the present invention is an angiopoietin, which in one embodiment, is Angiopoietin 1, Angiopoietin 3, Angiopoietin 4 or Angiopoietin 6.
  • endoglin is also known as CD105; EDG; HHT1; ORW; or ORW1.
  • endoglin is a TGFbeta co-receptor.
  • the CAR T-cells bind to an antigen associated with an infectious agent.
  • the infectious agent is Mycobacterium tuberculosis.
  • said Mycobacterium tuberculosis associated antigen is: Antigen 85B, Lipoprotein IpqH, ATP dependent helicase putative, uncharacterized protein Rv0476/MTO4941 precursor or uncharacterized protein Rvl334/MT1376 precursor.
  • the CAR binds to an antibody.
  • the CAR T-cell is an "antibody-coupled T-cell receptor" (ACTR).
  • ACTR antibody-coupled T-cell receptor
  • the CAR T-cell is a universal CAR T-cell.
  • the CAR T-cell having an antibody receptor is administered before, after, or at the same time as the antibody is administered and then binds to the antibody, bringing the T-cell in close proximity to the tumor or cancer.
  • the antibody is directed against a tumor cell antigen.
  • the antibody is directed against CD20.
  • the antibody is rituximab.
  • the antibody is Trastuzumab (Herceptin; Genentech): humanized IgGl, which is directed against ERBB2.
  • the antibody is Bevacizumab (Avastin; Genentech/Roche): humanized IgGl, which is directed against VEGF.
  • the antibody is Cetuximab (Erbitux; Bristol-Myers Squibb): chimeric human-murine IgGl, which is directed against EGFR.
  • the antibody is Panitumumab (Vectibix; Amgen): human IgG2, which is directed against EGFR.
  • the antibody is Ipilimumab (Yervoy; Bristol-Myers Squibb): IgGl, which is directed against CTLA4.
  • the antibody is Alemtuzumab (Campath; Genzyme): humanized IgGl, which is directed against CD52.
  • the antibody is Ofatumumab (Arzerra; Genmab): human IgGl, which is directed against CD20.
  • the antibody is Gemtuzumab ozogamicin (Mylotarg; Wyeth): humanized IgG4, which is directed against CD33.
  • the antibody is Brentuximab vedotin (Adcetris; Seattle Genetics): chimeric IgGl, which is directed against CD30.
  • the antibody is 90Y-labelled ibritumomab tiuxetan (Zevalin; IDEC Pharmaceuticals): murine IgGl, which is directed against CD20.
  • the antibody is 1311-labelled tositumomab (Bexxar; GlaxoSmithKline): murine IgG2, which is directed against CD20.
  • the antibody is Ramucirumab, which is directed against vascular endothelial growth factor receptor-2 (VEGFR-2).
  • the antibody is ramucirumab (Cyramza Injection, Eli Lilly and Company), blinatumomab (BLINCYTO, Amgen Inc.), pembrolizumab (KEYTRUDA, Merck Sharp & Dohme Corp.), obinutuzumab (GAZYVA, Genentech, Inc.; previously known as GA101), pertuzumab injection (PERJETA, Genentech, Inc.), or denosumab (Xgeva, Amgen Inc.).
  • the antibody is Basiliximab (Simulect; Novartis).
  • the antibody is Daclizumab (Zenapax; Roche).
  • the antibody to which the CAR T-cell is coupled is directed to a tumor or cancer antigen or a portion thereof, that is described herein and/or that is known in the art.
  • the antibody to which the CAR T-cell is couples is directed to a tumor- associated antigen.
  • the antibody to which the CAR T-cell is couples is directed to a tumor-associated antigen or a portion thereof that is an angiogenic factor.
  • a genetically modified TCR may be engineered to recognize any of the antigens described above to which a CAR binds.
  • a TCR T-cell binds to an antigen described above as a CAR T-cell binding target.
  • a TCR recognizes any antigen disclosed herein.
  • the antigen to which the TCR recognizes is a tumor or cancer antigen or a portion thereof, that is described herein and/or that is known in the art.
  • the TCR recognizes a tumor-associated antigen.
  • the TCR recognizes a tumor-associated antigen or a portion thereof that is an angiogenic factor.
  • DCs dendritic cells
  • DCs are antigen-producing and presenting cells of the mammalian immune system that process antigen material and present it on the cell surface to the T- cells of the immune system and are thereby capable of sensitizing T-cells to both new and recall antigens.
  • DCs are the most potent antigen-producing cells, acting as messengers between the innate and the adaptive immune systems. DC cells may be used, in one embodiment, to prime specific antitumor immunity through the generation of effector cells that attack and lyse tumors.
  • Dendritic cells are present in those tissues that are in contact with the external environment, such as the skin (where there is a specialized dendritic cell type called the Langerhans cell) and the inner lining of the nose, lungs, stomach and intestines. They can also be found in an immature state in the blood. Once activated, they migrate to the lymph nodes where they interact with T-cells and B cells to initiate and shape the adaptive immune response. At certain development stages, they grow branched projections, the dendrites that give the cell its name. Dendritic cells may be engineered to express particular tumor antigens.
  • the three signals that are required for T-cell activation are: (i) presentation of cognate antigen in self MHC molecules; (ii) costimulation by membrane-bound receptor-ligand pairs; and (iii) soluble factors to direct polarization of the ensuing immune response.
  • Dendritic cells (DCs) are able to provide all of the three signals required for T-cell activation making them an excellent cancer vaccine platform.
  • compositions comprising dendritic cells and an additional agent, wherein said additional agent comprises apoptotic cells, apoptotic supernatants, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • additional agent comprises apoptotic cells, apoptotic supernatants, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • a method of treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject comprising the step of administering dendritic cells and a composition comprising an additional agent, wherein said agent comprises apoptotic cells, apoptotic supernatants, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, to said subject.
  • a method as disclosed herein includes providing immune cells, such as NK cells, dendritic cells, TCR T-cells, or T-cells comprising engineered chimeric antigen receptors (CAR T-cells), with at least an additional agent to decrease toxic cytokine release or "cytokine release syndrome” (CRS) or "severe cytokine release syndrome” (sCRS) or "cytokine storm” that may occur in the subject.
  • CRS cytokine release syndrome
  • sCRS severe cytokine release syndrome
  • cytokine storm occurs as a result of administration of the immune cells.
  • the CRS, sCRS or cytokine storm is the result of a stimulus, condition, or syndrome separate from the immune cells (see below).
  • a cytokine storm, cytokine cascade, or hypercytokinemia is a more severe form of cytokine release syndrome.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or a composition comprising said apoptotic cells.
  • the additional agent for decreasing harmful cytokine release comprises an apoptotic cell supernatant or a composition comprising said supernatant.
  • the additional agent for decreasing harmful cytokine release comprises a CTLA-4 blocking agent.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and a CTLA-4 blocking agent.
  • the additional agent for decreasing harmful cytokine release comprises an alpha- 1 anti-trypsin or fragment thereof or analogue thereof.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and an alpha- 1 anti-trypsin or fragment thereof or analogue thereof.
  • the additional agent for decreasing harmful cytokine release comprises a tellurium-based compound.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and a tellurium-based compound.
  • the additional agent for decreasing harmful cytokine release comprises an immune modulating agent.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and an immune modulating agent.
  • the additional agent for decreasing harmful cytokine release comprises Treg cells.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and Treg cells.
  • decreasing toxic cytokine release or toxic cytokine levels comprises decreasing or inhibiting production of toxic cytokine levels in a subject, or inhibiting or reducing the incidence of cytokine release syndrome or a cytokine storm in a subject.
  • toxic cytokine levels are reduced during CRS or a cytokine storm.
  • decreasing or inhibiting the production of toxic cytokine levels comprises treating CRS or a cytokine storm.
  • decreasing or inhibiting the production of toxic cytokine levels comprises preventing CRS or a cytokine storm.
  • decreasing or inhibiting the production of toxic cytokine levels comprises alleviating CRS or a cytokine storm.
  • decreasing or inhibiting the production of toxic cytokine levels comprises ameliorating CRS or a cytokine storm.
  • the toxic cytokines comprise pro-inflammatory cytokines.
  • pro-inflammatory cytokines comprise IL-6.
  • pro-inflammatory cytokines comprise IL- ⁇ .
  • pro-inflammatory cytokines comprise TNF-a,
  • pro- inflammatory cytokines comprise IL-6, IL-1 ⁇ , or TNF-a, or any combination thereof.
  • cytokine release syndrome is characterized by elevated levels of several inflammatory cytokines and adverse physical reactions in a subject such as low blood pressure, high fever and shivering.
  • inflammatory cytokines comprise IL-6, IL- ⁇ , and TNF-a.
  • CRS is characterized by elevated levels of IL-6, IL- ⁇ , or TNF-a, or any combination thereof.
  • CRS is characterized by elevated levels of IL- 8, or IL-13, or any combination thereof.
  • a cytokine storm is characterized by increases in TNF-alpha, IFN-gamma, IL-1 beta, IL-2, IL-6, IL-8, IL-10, IL-13, GM-CSF, IL-5, fracktalkine, or a combination thereof or a subset thereof.
  • IL-6 comprises a marker of CRS or cytokine storm.
  • patients with larger tumor burdens have higher incidence and severity of cytokine release syndrome.
  • cytokines increased in CRS or a cytokine storm in humans and mice may comprise any combination of cytokines listed in Tables 1 and 2 below.
  • Table 1 Panel of Cytokines Increased in CRS or Cytokine Storm in Humans and/or Mice Human Mouse model (pre-clinical)
  • fibroblasts fibroblasts, keratinocytes,
  • adipocytes adipocytes, myocytes,
  • cytokines Flt-3L, Fractalkine, GM-CSF, IFN- ⁇ , IL- ⁇ , IL-2, IL-2Ra, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, and IL-13 of Table 1 are considered to be significant in CRS or cytokine storm.
  • IFN-a, IFN- ⁇ , IL-1, and IL-lRa of Table 1 appear to be important in CRS or cytokine storm.
  • M-CSF has unknown importance.
  • any cytokine listed in Table 1, or combination thereof may be used as a marker of CRS or cytokine storm.
  • T cells CCL3 platelets, endothelial cells, neutrophils, monocytes,
  • PAF ? CCL4 and macrophages, mesangial cells
  • lymphocytes lymphocytes
  • TGF- ⁇ * * CCL5 endothel, platelets ...
  • IL-15, IL-17, IL-18, IL-21, IL-22, IP-10, MCP-1, ⁇ - ⁇ , ⁇ - ⁇ , and TNF-a of Table 2 are considered to be significant in CRS or cytokine storm.
  • IL-27, MCP-3, PGE2, RANTES, TGF- ⁇ , TNF-aRl , and MIG of Table 2 appear to be important in CRS or cytokine storm.
  • IL-23 and IL-25 have unknown importance.
  • any cytokine listed in Table 2, or combination thereof may be used as a marker of CRS or cytokine storm.
  • mouse cytokines IL-10, IL- 1 ⁇ , IL-2, IP-10, IL-4, IL-5, IL-6, IFNa, IL-9, IL-13, IFN- ⁇ , IL-12p70, GM-CSF, TNF-a, MIP-la, ⁇ - ⁇ , IL-17 A, IL-15/IL-15R and IL-7 appear to be important in CRS or cytokine storm.
  • cytokine may encompass cytokines (e.g., interferon gamma (IFN- ⁇ ), granulocyte macrophage colony stimulating factor, tumor necrosis factor alpha), chemokines (e.g., MIP 1 alpha, MIP 1 beta, RANTES), and other soluble mediators of inflammation, such as reactive oxygen species and nitric oxide.
  • cytokines e.g., interferon gamma (IFN- ⁇ ), granulocyte macrophage colony stimulating factor, tumor necrosis factor alpha), chemokines (e.g., MIP 1 alpha, MIP 1 beta, RANTES), and other soluble mediators of inflammation, such as reactive oxygen species and nitric oxide.
  • IFN- ⁇ interferon gamma
  • chemokines e.g., MIP 1 alpha, MIP 1 beta, RANTES
  • other soluble mediators of inflammation such as reactive oxygen species and nitric oxide.
  • increased release of a particular cytokine does not a priori mean that the particular cytokine is part of a cytokine storm.
  • an increase of at least one cytokine is not the result of a cytokine storm or CRS.
  • CAR T-cells may be the source of increased levels of a particular cytokine or group of cytokines.
  • cytokine release syndrome is characterized by any or all of the following symptoms: Fever with or without rigors, malaise, fatigue, anorexia, myalgias, arthalgias, nausea, vomiting, headache Skin Rash, Nausea, vomiting, diarrhea, Tachypnea, hypoxemia Cardiovascular Tachycardia, widened pulse pressure, hypotension, increased cardiac output (early), potentially diminished cardiac output (late), Elevated D-dimer, hypofibrinogenemia with or without bleeding, Azotemia Hepatic Transaminitis, hyperbilirubinemia, Headache, mental status changes, confusion, delirium, word finding difficulty or frank aphasia, hallucinations, tremor, dymetria, altered gait, seizures.
  • a cytokine storm is characterized by IL-2 release and lymphoproliferation.
  • a cytokine storm is characterized by increases in cytokines released by CAR T-cells.
  • a cytokine storm is characterized by increases in cytokines released by cells other than CAR T-cells.
  • cytokine storm leads to potentially life-threatening complications including cardiac dysfunction, adult respiratory distress syndrome, neurologic toxicity, renal and/or hepatic failure, and disseminated intravascular coagulation.
  • CRS cytokine release syndrome
  • cytokine storm the characteristics of a cytokine release syndrome (CRS) or cytokine storm are estimated to occur a few days to several weeks following the trigger for the CRS or cytokine storm.
  • CAR T-cells are a trigger for CRS or a cytokine storm.
  • a trigger for CRS or a cytokine storm is not CAR T-cells.
  • measurement of cytokine levels or concentration, as an indicator of cytokine storm may be expressed as -fold increase, per cent (%) increase, net increase or rate of change in cytokine levels or concentration.
  • absolute cytokine levels or concentrations above a certain level or concentration may be an indication of a subject undergoing or about to experience a cytokine storm.
  • absolute cytokine levels or concentration at a certain level or concentration for example a level or concentration normally found in a control subject not undergoing CAR-T cell therapy, may be an indication of a method for inhibiting or reducing the incidence of a cytokine storm in a subject undergoing CAR T-cell.
  • cytokine level may encompass a measure of concentration, a measure of fold change, a measure of percent (%) change, or a measure of rate change.
  • cytokine level may encompass a measure of concentration, a measure of fold change, a measure of percent (%) change, or a measure of rate change.
  • IL-6 levels may be used as a common measure of cytokine storm and/or as a common measure of the effectiveness of a treatment for cytokine storms.
  • cytokines may be used as markers of a cytokine storm, for example TNF-a, IB-la, IL-8, IL-13, or INF- ⁇ .
  • assay methods for measuring cytokines are well known in the art. A skilled artisan would appreciate that methods affecting a cytokine storm may similarly affect cytokine release syndrome.
  • disclosed herein is a method of decreasing or inhibiting cytokine production in a subject experiencing cytokine release syndrome or a cytokine storm. In another embodiment, disclosed herein is a method of decreasing or inhibiting cytokine production in a subject vulnerable to experiencing cytokine release syndrome or a cytokine storm. In another embodiment, methods disclosed herein decrease or inhibit cytokine production in a subject experiencing cytokine release syndrome or a cytokine storm, wherein production of any cytokine or group of cytokines listed in Tables 1 and/or 2 is decreased or inhibited. In another embodiment, cytokine IL-6 production is decreased or inhibited.
  • cytokine IL-betal production is decreased or inhibited.
  • cytokine IL-8 production is decreased or inhibited.
  • cytokine IL-13 production is decreased or inhibited.
  • cytokine TNF-alpha production is decreased or inhibited.
  • cytokines IL-6 production, IL-lbeta production, or TNF-alpha production, or any combination thereof is decreased or inhibited.
  • cytokine release syndrome is graded.
  • Grade 1 describes cytokine release syndrome in which symptoms are not life threatening and require symptomatic treatment only, e.g., fever, nausea, fatigue, headache, myalgias, malaise.
  • Grade 2 symptoms require and respond to moderate intervention, such as oxygen, fluids or vasopressor for hypotension.
  • Grade 3 symptoms require and respond to aggressive intervention.
  • Grade 4 symptoms are life-threatening symptoms and require ventilator and patients display organ toxicity.
  • a cytokine storm is characterized by IL-6 and interferon gamma release.
  • a cytokine storm is characterized by release of any cytokine or combination thereof, listed in Tables 1 and 2.
  • a cytokine storm is characterized by release of any cytokine or combination thereof, known in the art.
  • symptoms onset begins minutes to hours after the infusion begins. In another embodiment, symptoms coincide with peak cytokine levels.
  • a method of inhibiting or reducing the incidence of a cytokine release syndrome (CRS) or a cytokine storm in a subject undergoing CAR T-cell cancer therapy comprises administering an apoptotic cell population or an apoptotic cell supernatant or compositions thereof.
  • the apoptotic cell population or an apoptotic cell supernatant or compositions thereof may aid the CAR T-cell therapy.
  • the apoptotic cell population or an apoptotic cell supernatant or compositions thereof may aid in the inhibition or reducing the incidence of the CRS or cytokine storm.
  • the apoptotic cell population or an apoptotic cell supernatant or compositions thereof may aid in treating the CRS or cytokine storm. In another embodiment, the apoptotic cell population or an apoptotic cell supernatant or compositions thereof may aid in preventing the CRS or cytokine storm. In another embodiment, the apoptotic cell population or an apoptotic cell supernatant or compositions thereof may aid in ameliorating the CRS or cytokine storm. In another embodiment, the apoptotic cell population or an apoptotic cell supernatant or compositions thereof may aid in alleviating the CRS or cytokine storm.
  • a method of inhibiting or reducing the incidence of a cytokine release syndrome (CRS) or a cytokine storm in a subject undergoing CAR T-cell cancer therapy, and being administered an apoptotic cell population or an apoptotic cell supernatant or compositions thereof comprises administering an additional agent.
  • the additional agent may aid the CAR T-cell therapy.
  • the additional agent may aid in the inhibition or reducing the incidence of the CRS or cytokine storm.
  • the additional agent may aid in treating the CRS or cytokine storm.
  • the additional agent may aid in preventing the CRS or cytokine storm.
  • the additional agent may aid in ameliorating the CRS or cytokine storm.
  • the additional agent may aid in alleviating the CRS or cytokine storm.
  • a method of inhibiting or reducing the incidence of a cytokine release syndrome (CRS) or a cytokine storm in a subject undergoing CAR T-cell cancer therapy comprises administering an additional agent.
  • the additional agent may aid the CAR T- cell therapy.
  • a method of inhibiting or reducing the incidence of a cytokine release syndrome (CRS) or a cytokine storm in a subject undergoing TCR T-cell cancer therapy comprises administering an additional agent.
  • the additional agent may aid the TCR T-cell therapy.
  • a method of inhibiting or reducing the incidence of a cytokine release syndrome (CRS) or a cytokine storm in a subject undergoing comprises administering an additional agent.
  • the additional agent may aid the .
  • a method of inhibiting or reducing the incidence of a cytokine release syndrome (CRS) or a cytokine storm in a subject undergoing NK cell therapy comprises administering an additional agent.
  • the additional agent may aid the NK cell therapy.
  • the additional agent may aid in the inhibition or reducing the incidence of the CRS or cytokine storm. In another embodiment, the additional agent may aid in treating the CRS or cytokine storm. In another embodiment, the additional agent may aid in preventing the CRS or cytokine storm. In another embodiment, the additional agent may aid in ameliorating the CRS or cytokine storm. In another embodiment, the additional agent may aid in alleviating the CRS or cytokine storm.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or a composition comprising said apoptotic cells.
  • the additional agent for decreasing harmful cytokine release comprises an apoptotic cell supernatant or a composition comprising said supernatant.
  • the additional agent for decreasing harmful cytokine release comprises a CTLA-4 blocking agent.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and a CTLA-4 blocking agent.
  • the additional agent for decreasing harmful cytokine release comprises an alpha- 1 anti-trypsin or fragment thereof or analogue thereof.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and an alpha- 1 anti-trypsin or fragment thereof or analogue thereof.
  • the additional agent for decreasing harmful cytokine release comprises a tellurium-based compound.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and a tellurium-based compound.
  • the additional agent for decreasing harmful cytokine release comprises an immune modulating agent.
  • the additional agent for decreasing harmful cytokine release comprises apoptotic cells or apoptotic cell supernatants or compositions thereof, and an immune modulating agent.
  • compositions and methods as disclosed herein utilize combination therapy of CAR T-cells with one or more CTLA-4-blocking agents such as Ipilimumab.
  • compositions and methods as disclosed herein utilize combined therapy comprising apoptotic cells, CAR T-cells, and one or more CTLA-4-blocking agents.
  • compositions and methods as disclosed herein utilize combination therapy of TCR T-cells with one or more CTLA-4-blocking agents such as Ipilimumab.
  • compositions and methods as disclosed herein utilize combined therapy comprising apoptotic cells, TCR T-cells, and one or more CTLA-4-blocking agents.
  • compositions and methods as disclosed herein utilize combination therapy of dendritic cells with one or more CTLA-4-blocking agents such as Ipilimumab.
  • compositions and methods as disclosed herein utilize combined therapy comprising apoptotic cells, dendritic cells, and one or more CTLA-4- blocking agents.
  • compositions and methods as disclosed herein utilize combination therapy of NK cells with one or more CTLA-4-b locking agents such as Ipilimumab.
  • compositions and methods as disclosed herein utilize combined therapy comprising apoptotic cells, NK cells, and one or more CTLA-4-blocking agents.
  • CTLA-4 is a potent inhibitor of T-cell activation that helps to maintain self-tolerance.
  • administration of an anti-CTLA-4 blocking agent, which in another embodiment, is an antibody produces a net effect of T-cell activation.
  • compositions and methods as disclosed herein comprise B cell aplasia or tumor lysis syndrome (TLS).
  • TLS tumor lysis syndrome
  • a method of inhibiting or reducing the incidence of a cytokine release syndrome (CRS) or a cytokine storm in a subject undergoing CAR T-cell cancer therapy does not affect the efficacy of the CAR T-cell therapy.
  • a method of inhibiting or reducing the incidence of CRS or a cytokine storm in a subject undergoing CAR T-cell cancer therapy does reduce the efficacy of the CAR T-cells therapy by more than about 5%.
  • a method of inhibiting or reducing the incidence of CRS or a cytokine storm in a subject undergoing CAR T-cell cancer therapy does reduce the efficacy of the CAR T-cells therapy by more than about 10%.
  • a method of inhibiting or reducing the incidence of CRS or a cytokine storm in a subject undergoing CAR T-cell cancer therapy does reduce the efficacy of the CAR T-cells therapy by more than about 15%. In another embodiment, a method of inhibiting or reducing the incidence of CRS or a cytokine storm in a subject undergoing CAR T-cell cancer therapy, does reduce the efficacy of the CAR T-cells therapy by more than about 20%.
  • cytotoxicity can be quantified using a cell culture-based assay such as the cytotoxic assays described in the Examples.
  • Cytotoxicity assays can employ dyes that preferentially stain the DNA of dead cells.
  • fluorescent and luminescent assays that measure the relative number of live and dead cells in a cell population can be used.
  • protease activities serve as markers for cell viability and cell toxicity, and a labeled cell permeable peptide generates fluorescent signals that are proportional to the number of viable cells in the sample.
  • a cytotoxicity assay may use 7-AAD in a flow cytometry analysis. Kits for various cytotoxicity assays are commercially available from manufacturers such as Promega, Abeam, and Life Technologies.
  • a measure of cytotoxicity may be qualitative. In another embodiment, a measure of cytotoxicity may be quantitative. In a further embodiment a measure of cytotoxicity may be related to the change in expression of a cytotoxic cytokine. In another embodiment, a measure of cytotoxicity may be determined by survival curve and tumor load in bone marrow and liver.
  • the methods as disclosed herein comprise an additional step that is useful in overcoming rejection of allogeneic donor cells.
  • the methods comprise the step of full or partial lymphodepletion prior to administration of the CAR T-cells, which in one embodiment, are allogeneic CAR T-cells.
  • the lymphodepletion is adjusted so that it delays the host versus graft reaction for a period sufficient to allow said allogeneic T-cells to attack the tumor to which they are directed, but to an extent insufficient to require rescue of the host immune system by bone marrow transplantation.
  • agents that delay egression of the allogeneic T-cells from lymph nodes such as 2-amino-2-[2-(4- octylphenyl)ethyl]propane-l ,3-diol (FTY720), 5-[4-phenyl-5-(trifiuoromethyl)thiophen-2-yl]-3-[3- (trifiuoromethyl)pheny- l]l ,2,4-oxadiazole (SEW2871), 3-(2-(-hexylphenylamino)-2- oxoethylamino)propanoic acid (W123), 2-ammonio-4-(2-chloro-4-(3-phenoxyphenylthio)phenyl)- 2-(hydroxymethyl)but-yl hydrogen phosphate (KRP-203 phosphate) or other agents known in the art, may be used as part of the compositions and methods as disclosed herein to allow the use of allogeneic C
  • cytokine release occurs between a few days to 2 weeks after administration of immune therapy such as CAR T-cell therapy.
  • immune therapy such as CAR T-cell therapy.
  • hypotension and other symptoms follow the cytokine release, i.e. from few days to few weeks. Therefore, in one embodiment, apoptotic cells or the apoptotic cell supernatant are administered to subjects at the same time as immune therapy as prophylaxis.
  • apoptotic cells or supernatant are administered to subjects 2-3 days after administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 7 days after administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 10 days after administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 14 days after administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 2-14 days after administration of immune therapy.
  • apoptotic cells or apoptotic cell supernatant are administered to subjects 2-3 hours after administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 7 hours after administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 10 hours after administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 14 hours after administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 2-14 hours after administration of immune therapy.
  • apoptotic cells or the apoptotic cell supernatant are administered to subjects prior to immune therapy as prophylaxis.
  • apoptotic cells or supernatant are administered to subjects 1 day before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 2-3 days before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 7 days before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 10 days before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 14 days before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 2-14 days before administration of immune therapy.
  • apoptotic cells or apoptotic cell supernatant are administered to subjects 2-3 hours before administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 7 hours before administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 10 hours before administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 14 hours before administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 2-14 hours before administration of immune therapy.
  • apoptotic cells or apoptotic cell supernatant may be administered therapeutically, once cytokine release syndrome has occurred. In one embodiment, apoptotic cells or supernatant may be administered once cytokine release leading up to or attesting to the beginning of cytokine release syndrome is detected. In one embodiment, apoptotic cells or supernatant can terminate the increased cytokine levels, or the cytokine release syndrome, and avoid its sequelae.
  • apoptotic cells or apoptotic cell supernatant may be administered therapeutically, at multiple time points.
  • administration of apoptotic cells or apoptotic cell supernatant is at least at two time points described herein.
  • administration of apoptotic cells or apoptotic cell supernatant is at least at three time points described herein.
  • administration of apoptotic cells or apoptotic cell supernatant is prior to CRS or a cytokine storm, and once cytokine release syndrome has occurred, and any combination thereof.
  • the chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy and the apoptotic cell therapy or supernatant are administered together.
  • the CAR T-cell therapy is administered after the apoptotic cell therapy or supernatant.
  • the CAR T-cell therapy is administered prior to the apoptotic cell therapy or supernatant.
  • apoptotic cell therapy or supernatant is administered approximately 2-3 weeks after the CAR T-cell therapy.
  • apoptotic cell therapy or supernatant is administered approximately 6-7 weeks after the CAR T-cell therapy.
  • apoptotic cell therapy or supernatant is administered approximately 9 weeks after the CAR T-cell therapy.
  • apoptotic cell therapy is administered up to several months after CAR T-cell therapy.
  • apoptotic cells or the apoptotic cell supernatant are administered to subjects at the same time as immune therapy as prophylaxis.
  • apoptotic cells or supernatant are administered to subjects 2-3 days after administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 7 days after administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 10 days after administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 14 days after administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 2-14 days after administration of immune therapy.
  • apoptotic cells or apoptotic cell supernatant are administered to subjects 2-3 hours after administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 7 hours after administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 10 hours after administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 14 hours after administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 2-14 hours after administration of immune therapy.
  • apoptotic cells or the apoptotic cell supernatant are administered to subjects prior to immune therapy as prophylaxis.
  • apoptotic cells or supernatant are administered to subjects 1 day before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 2-3 days before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 7 days before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 10 days before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 14 days before administration of immune therapy.
  • apoptotic cells or supernatant are administered to subjects 2-14 days before administration of immune therapy.
  • apoptotic cells or apoptotic cell supernatant are administered to subjects 2-3 hours before administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 7 hours before administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 10 hours before administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 14 hours before administration of immune therapy. In another embodiment, apoptotic cells or supernatant are administered to subjects 2-14 hours before administration of immune therapy.
  • apoptotic cells or apoptotic cell supernatant may be administered therapeutically, once cytokine release syndrome has occurred. In one embodiment, apoptotic cells or supernatant may be administered once cytokine release leading up to or attesting to the beginning of cytokine release syndrome is detected. In one embodiment, apoptotic cells or supernatant can terminate the increased cytokine levels, or the cytokine release syndrome, and avoid its sequelae.
  • apoptotic cells or apoptotic cell supernatant may be administered therapeutically, at multiple time points.
  • administration of apoptotic cells or apoptotic cell supernatant is at least at two time points described herein.
  • administration of apoptotic cells or apoptotic cell supernatant is at least at three time points described herein.
  • administration of apoptotic cells or apoptotic cell supernatant is prior to CRS or a cytokine storm, and once cytokine release syndrome has occurred, and any combination thereof.
  • the chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy and the apoptotic cell therapy or supernatant are administered together.
  • the CAR T-cell therapy is administered after the apoptotic cell therapy or supernatant.
  • the CAR T-cell therapy is administered prior to the apoptotic cell therapy or supernatant.
  • apoptotic cell therapy or supernatant is administered approximately 2-3 weeks after the CAR T-cell therapy.
  • apoptotic cell therapy or supernatant is administered approximately 6-7 weeks after the CAR T-cell therapy.
  • apoptotic cell therapy or supernatant is administered approximately 9 weeks after the CAR T-cell therapy.
  • apoptotic cell therapy is administered up to several months after CAR T-cell therapy.
  • an additional agent is administered to subjects at the same time as immune therapy as prophylaxis.
  • the additional agent comprises apoptotic cells, an apoptotic supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, of a tellurium-based compound, or an immune-modulating compounds, or any combination thereof.
  • the additional agent is administered to subjects 2-3 days after administration of immune therapy.
  • the additional agent is administered to subjects 7 days after administration of immune therapy.
  • the additional agent is administered to subjects 10 days after administration of immune therapy.
  • the additional agent is administered to subjects 14 days after administration of immune therapy.
  • the additional agent is administered to subjects 2-14 days after administration of immune therapy.
  • the additional agent is administered to subjects 2-3 hours after administration of immune therapy. In another embodiment, the additional agent is administered to subjects 7 hours after administration of immune therapy. In another embodiment the additional agent is administered to subjects 10 hours after administration of immune therapy. In another embodiment, the additional agent is administered to subjects 14 hours after administration of immune therapy. In another embodiment, the additional agent is administered to subjects 2-14 hours after administration of immune therapy.
  • the additional agent is administered to subjects prior to immune therapy as prophylaxis. In another embodiment, the additional agent is administered to subjects 1 day before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 2-3 days before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 7 days before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 10 days before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 14 days before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 2-14 days before administration of immune therapy.
  • the additional agent is administered to subjects 2-3 hours before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 7 hours before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 10 hours before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 14 hours before administration of immune therapy. In another embodiment, the additional agent is administered to subjects 2-14 hours before administration of immune therapy.
  • the additional agent is administered therapeutically, once cytokine release syndrome has occurred. In one embodiment, the additional agent is administered once cytokine release leading up to or attesting to the beginning of cytokine release syndrome is detected. In one embodiment, the additional agent can terminate the increased cytokine levels, or the cytokine release syndrome, and avoid its sequelae.
  • the additional agent is administered therapeutically, at multiple time points. In another embodiment, administration of the additional agent is at least at two time points described herein. In another embodiment, administration of the additional agent is at least at three time points described herein. In another embodiment, administration of the additional agent is prior to CRS or a cytokine storm, and once cytokine release syndrome has occurred, and any combination thereof.
  • the chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy and the additional agent is administered together.
  • the CAR T-cell therapy is administered the additional agent.
  • the CAR T-cell therapy is administered prior to the additional agent.
  • the additional agent is administered approximately 2-3 weeks after the CAR T-cell therapy.
  • the additional agent is administered approximately 6-7 weeks after the CAR T-cell therapy.
  • the additional agent is administered approximately 9 weeks after the CAR T-cell therapy.
  • the additional agent is administered up to several months after CAR T-cell therapy.
  • CAR T-cells are heterologous to the subject.
  • CAR T-cells are derived from one or more donors.
  • CAR T-cells are derived from one or more bone marrow donors.
  • CAR T-cells are derived from one or more blood bank donations.
  • the donors are matched donors.
  • CAR T-cells are universal allogeneic CAR T-cells.
  • CAR T- cells are syngeneic CAR T-cells.
  • CAR T-cells are from unmatched third party donors.
  • CAR T-cells are from pooled third party donor T-cells.
  • the donor is a bone marrow donor. In another embodiment, the donor is a blood bank donor.
  • CAR T-cells of the compositions and methods as disclosed herein comprise one or more MHC unrestricted tumor-directed chimeric receptors.
  • non-autologous T-cells may be engineered or administered according to protocols known in the art to prevent or minimize autoimmune reactions, such as described in U.S. Patent Application No. 20130156794, which is incorporated herein by references in its entirety.
  • CAR T-cells are autologous to the subject. In one embodiment, the patient's own cells are used. In this embodiment, if the patient's own cells are used, then the CAR T-cell therapy is administered after the apoptotic cell therapy.
  • apoptotic cells are heterologous to the subject.
  • apoptotic cells are derived from one or more donors.
  • apoptotic cells are derived from one or more bone marrow donors.
  • apoptotic cells are derived from one or more blood bank donations.
  • the donors are matched donors.
  • apoptotic cells are from unmatched third party donors.
  • apoptotic cells are universal allogeneic apoptotic cells.
  • apoptotic cells are from a syngeneic donor.
  • apoptotic cells are from pooled third party donor cells.
  • the donor is a bone marrow donor.
  • the donor is a blood bank donor.
  • apoptotic cells are autologous to the subject. In this embodiment, the patient's own cells are used.
  • the therapeutic mononuclear-enriched cell preparation disclosed herein or the apoptotic cell supernatant is administered to the subject systemically. In another embodiment, administration is via the intravenous route. Alternately, the therapeutic mononuclear enriched cell or supernatant may be administered to the subject according to various other routes, including, but not limited to, the parenteral, intraperitoneal, intra-articular, intramuscular and subcutaneous routes. Each possibility represents a separate embodiment as disclosed herein.
  • the therapeutic mononuclear-enriched cell preparation disclosed herein or the additional agent is administered to the subject systemically. In another embodiment, administration is via the intravenous route. Alternately, the therapeutic mononuclear enriched cell or the additional agent may be administered to the subject according to various other routes, including, but not limited to, the parenteral, intraperitoneal, intra-articular, intramuscular and subcutaneous routes. Each possibility represents a separate embodiment as disclosed herein.
  • the preparation is administered in a local rather than systemic manner, for example, via injection of the preparation directly into a specific region of a patient's body.
  • a specific region comprises a tumor or cancer.
  • the therapeutic mononuclear enriched cells or supernatant are administered to the subject suspended in a suitable physiological buffer, such as, but not limited to, saline solution, PBS, HBSS, and the like.
  • a suitable physiological buffer such as, but not limited to, saline solution, PBS, HBSS, and the like.
  • the suspension medium may further comprise supplements conducive to maintaining the viability of the cells.
  • the additional agent is administered to the subject suspended in a suitable physiological buffer, such as, but not limited to, saline solution, PBS, HBSS, and the like.
  • the pharmaceutical composition is administered intravenously. According to another embodiment, the pharmaceutical composition is administered in a single dose. According to alternative embodiments the pharmaceutical composition is administered in multiple doses. According to another embodiment, the pharmaceutical composition is administered in two doses. According to another embodiment, the pharmaceutical composition is administered in three doses. According to another embodiment, the pharmaceutical composition is administered in four doses. According to another embodiment, the pharmaceutical composition is administered in five or more doses. According to some embodiments, the pharmaceutical composition is formulated for intravenous injection.
  • any appropriate method of providing modified CAR-expressing immune cells to a subject can be used for methods described herein.
  • methods for providing cells to a subject comprise hematopoietic cell transplantation (HCT), infusion of donor-derived NK cells into cancer patients or a combination thereof.
  • HCT hematopoietic cell transplantation
  • a method of inhibiting or reducing the incidence of cytokine release syndrome or cytokine storm in a subject undergoing chimeric antigen receptor- expressing T-cell (CAR T-cell) therapy comprising the step of administering a composition comprising apoptotic cells to said subject.
  • CAR T-cell chimeric antigen receptor- expressing T-cell
  • a method of inhibiting or reducing the incidence of cytokine release syndrome or cytokine storm in a subject undergoing chimeric antigen receptor- expressing T-cell (CAR T-cell) therapy comprising the step of administering an apoptotic cell supernatant, such as an apoptotic cell-phagocyte supernatant, to said subject.
  • CAR T-cell chimeric antigen receptor- expressing T-cell
  • a method of inhibiting or reducing the incidence of cytokine release syndrome or cytokine storm in a subject undergoing chimeric antigen receptor- expressing T-cell (CAR T-cell) therapy comprising the step of administering an at least one additional agent to said subject.
  • CAR T-cell chimeric antigen receptor- expressing T-cell
  • a CAR T-cell therapy comprises administering a composition disclosed herein comprising CAR T-cells and either apoptotic cells or an apoptotic cell supernatant, or another or combination of additional agents as disclosed herein.
  • a CAR T-cell therapy comprises administering a composition disclosed herein comprising CAR T- cells and a composition comprising either apoptotic cells or an apoptotic cell supernatant, or an additional agent or combination thereof as disclosed herein..
  • a method of decreasing or inhibiting cytokine production in a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to cytokine release syndrome or cytokine storm comprising the step of administering a composition comprising apoptotic cells or an apoptotic supernatant to said subject, wherein said administering decreases or inhibits cytokine production in said subject.
  • decrease or inhibition of cytokine production is compared with a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to cytokine release syndrome or cytokine storm and not administered apoptotic cells or an apoptotic supernatant.
  • methods for decreasing or inhibiting cytokine production decrease or inhibit pro-inflammatory cytokine production. In another embodiment, methods for decreasing or inhibiting cytokine production decrease or inhibit production of at least one pro-inflammatory cytokine. In another embodiment, methods for decreasing or inhibiting cytokine production decrease or inhibit production of at least cytokine IL-6. In another embodiment, methods for decreasing or inhibiting cytokine production decrease or inhibit production of at least cytokine IL-lbeta. In another embodiment, methods for decreasing or inhibiting cytokine production decrease or inhibit production of at least cytokine TNF-alpha.
  • methods disclosed herein for decreasing or inhibiting cytokine production result in reduction or inhibition of production of cytokines IL-6, IL- ⁇ , or TNF-a, or any combination in said subject compared with a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to cytokine release syndrome or cytokine storm and not administered apoptotic cells or an apoptotic supernatant.
  • Cancers or tumors may also affect the absolute level of cytokines including proinflammatory cytokines.
  • the level of tumor burden in a subject may affect cytokine levels, particularly proOinflammatory cytokines.
  • cytokine levels particularly proOinflammatory cytokines.
  • a skilled artisan would appreciate that the phrase "decrease or inhibit” or grammatical variants thereof may encompass fold decrease or inhibition of cytokine production, or a net decrease or inhibition of cytokine production, or percent (%) decrease or inhibition, or may encompass a rate of change of decrease or inhibition of a cytokine production.
  • a method of decreasing or inhibiting cytokine production in a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to cytokine release syndrome or cytokine storm comprising the step of administering apoptotic cells or a composition comprising apoptotic cells to said subject.
  • a method of decreasing or inhibiting cytokine production in a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to cytokine release syndrome or cytokine storm comprising the step of administering an apoptotic cell supernatant, such as an apoptotic cell-phagocyte supernatant, or a composition comprising said supernatant to said subject.
  • an apoptotic cell supernatant such as an apoptotic cell-phagocyte supernatant, or a composition comprising said supernatant to said subject.
  • a method of decreasing or inhibiting cytokine production in a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to cytokine release syndrome or cytokine storm comprising the step of administering an apoptotic cell supernatant, such as an additional agent selected from the group comprising apoptotic cells, an apoptotic supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or a composition comprising said supernatant to said subject.
  • an apoptotic cell supernatant such as an additional agent selected from the group comprising apoptotic cells, an apoptotic supernatant, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or a composition comprising
  • an infection causes the cytokine release syndrome or cytokine storm in the subject.
  • the infection is an influenza infection.
  • the influenza infection is HlNl.
  • the influenza infection is an H5N1 bird flu.
  • the infection is severe acute respiratory syndrome (SARS).
  • the subject has Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis (HLH).
  • the infection is sepsis.
  • the sepsis is gram- negative.
  • the infection is malaria.
  • the infection is an Ebola virus infection.
  • the infection is variola virus.
  • the infection is a systemic Gram-negative bacterial infection.
  • the infection is Jarisch-Herxheimer syndrome.
  • the cause of the cytokine release syndrome or cytokine storm in a subject is hemophagocytic lymphohistiocytosis (HLH).
  • HLH is sporadic HLH.
  • HLH is macrophage activation syndrome (MAS).
  • MAS macrophage activation syndrome
  • the cause of the cytokine release syndrome or cytokine storm in a subject is MAS.
  • the cause of the cytokine release syndrome or cytokine storm in a subject is chronic arthritis.
  • the cause of the cytokine release syndrome or cytokine storm in a subject is systemic Juvenile Idiopathic Arthritis (sJIA), also known as Still' s Disease.
  • the cause of the cytokine release syndrome or cytokine storm in a subject is Cryopyrin-associated Periodic Syndrome (CAPS).
  • CAPS comprises Familial Cold Auto-inflammatory Syndrome (FCAS), also known as Familial Cold Urticaria (FCU).
  • FCAS Familial Cold Auto-inflammatory Syndrome
  • MWS Muckle-Well Syndrome
  • CAPS comprises Chronic Infantile Neurological Cutaneous and Articular (CINCA) Syndrome.
  • CAPS comprises FCAS, FCU, MWS, or CINCA Syndrome, or any combination thereof.
  • the cause of the cytokine release syndrome or cytokine storm in a subject is FCAS.
  • the cause of the cytokine release syndrome or cytokine storm in a subject is FCU. In another embodiment, the cause of the cytokine release syndrome or cytokine storm in a subject is MWS. In another embodiment, the cause of the cytokine release syndrome or cytokine storm in a subject is CINCA Syndrome. In still another embodiment, the cause of the cytokine release syndrome or cytokine storm in a subject is FCAS, FCU, MWS, or CINCA Syndrome, or any combination thereof.
  • the cause of the cytokine release syndrome or cytokine storm in a subject is a cryopyrinopathy comprising inherited or de novo gain of function mutations in the NLRP3 gene, also known as the CIAS I gene.
  • the cause of the cytokine release syndrome or cytokine storm in a subject is a hereditary auto-inflammatory disorder.
  • the trigger for the release of inflammatory cytokines is a lipopolysaccharide (LPS), Gram-positive toxins, fungal toxins, glycosylphosphatidylinositol (GPI) or modulation of RIG- 1 gene expression.
  • LPS lipopolysaccharide
  • GPI glycosylphosphatidylinositol
  • the subject experiencing cytokine release syndrome or cytokine storm does not have an infectious disease.
  • the subject has acute pancreatitis.
  • the subject has tissue injury, which in on embodiment, is severe burns or trauma.
  • the subject has acute respiratory distress syndrome.
  • the subject has cytokine release syndrome or cytokine storm secondary to drug use.
  • the subject has cytokine release syndrome or cytokine storm secondary to toxin inhalation.
  • the subject has cytokine release syndrome or cytokine storm secondary to receipt of immunotherapy, which in one embodiment is immunotherapy with superagonistic CD28-specific monoclonal antibodies (CD28SA).
  • CD28SA is TGN1412.
  • the immunotherapy is CAR T-cell therapy.
  • the immunotherapy is .
  • apoptotic cells or supernatant or a CTLA-4 blocking agent, an alpha- 1 anti- trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof may be used to control cytokine release syndrome or cytokine storm that results from administration of a pharmaceutical composition.
  • the pharmaceutical composition is oxaliplatin, cytarabine, lenalidomide, or a combination thereof.
  • apoptotic cells or the supernatant or a CTLA-4 blocking agent, an alpha- 1 anti- trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof may be used to control cytokine release syndrome or cytokine storm that results from administration of an antibody.
  • the antibody is monoclonal.
  • the antibody is polyclonal.
  • the antibody is rituximab.
  • the antibody is Orthoclone OKT3 (muromonab- CD3).
  • the antibody is alemtuzumab, tosituzumab, CP-870,893, LO- CD2a/BTI-322 or TGN1412.
  • examples of diseases for which control of inflammatory cytokine production can be beneficial include cancers, allergies, any type of infection, toxic shock syndrome, sepsis, any type of autoimmune disease, arthritis, Crohn's disease, lupus, psoriasis, or any other disease for which the hallmark feature is toxic cytokine release that causes deleterious effects in a subject.
  • Alpha- 1 -antitrypsin is a circulating 52-kDa glycoprotein that is produced mainly by the liver.
  • AAT is primarily known as a serine protease inhibitor and is encoded by the gene SERPINA1.
  • SERPINA1 serine protease inhibitor
  • AAT inhibits neutrophil elastase, and inherited deficiency in circulating AAT results in lung-tissue deterioration and liver disease. Serum AAT concentrations in healthy individuals increase twofold during inflammation
  • AAT AAT-associated hypertension
  • AAT human serum derived alpha- 1 -anti- trypsin
  • PBMC peripheral blood mononuclear cells
  • AAT reduces in vitro IL-l
  • AAT was shown to inhibit LPS- induced acute lung injury in experimental models. Recently, AAT was shown to reduce the size of infarct and the severity of heart failure in a mouse model of acute myocardial ischemia-reperfusion injury.
  • AAT monotherapy with clinical-grade human AAT (hAAT) reduced circulating proinflammatory cytokines, diminished Graft vs Host Disease (GvHD) severity, and prolonged animal survival after experimental allogeneic bone marrow transfer (Tawara et al., Proc Natl Acad Sci U S A. 2012 Jan 10;109(2):564-9), incorporated herein by reference.
  • AAT treatment reduced the expansion of alloreactive T effector cells but enhanced the recovery of T regulatory T-cells, (Tregs) thus altering the ratio of donor T effector to T regulatory cells in favor of reducing the pathological process.
  • AAT suppressed LPS-induced in vitro secretion of proinflammatory cytokines such as TNF-a and IL- ⁇ , enhanced the production of the anti- inflammatory cytokine IL-10, and impaired NF- ⁇ translocation in the host dendritic cells.
  • proinflammatory cytokines such as TNF-a and IL- ⁇
  • impaired NF- ⁇ translocation in the host dendritic cells Marcondes, Blood. 2014 (Oct 30;124(18):2881-91) incorporated herein by reference show that treatment with AAT not only ameliorated GvHD but also preserved and perhaps even enhanced the graft vs leukemia (GVL) effect.
  • VTL graft vs leukemia
  • compositions comprising chimeric antigen receptor-expressing T-cells (CAR T-cells) and Alpha- 1 -antitrypsin (AAT).
  • CAR T-cells and Alpha- 1 -antitrypsin (AAT) are in separate compositions.
  • AAT comprises a full length AAT or a functional fragment thereof.
  • AA comprises an analogue of a full length AAT or a functional fragment thereof.
  • a composition comprising AAT further comprises apoptotic cells or an apoptotic cell supernatant.
  • a method of treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject comprising the step of administering chimeric antigen receptor-expressing T-cells (CAR T-cells) and a composition comprising Alpha- 1 -antitrypsin (AAT) to said subject.
  • the method further comprises apoptotic cells or an apoptotic cell supernatant.
  • a method of inhibiting or reducing the incidence of cytokine release syndrome or cytokine storm in a subject undergoing chimeric antigen receptor- expressing T-cell (CAR T-cell) therapy comprising the step of administering a composition comprising Alpha- 1 -antitrypsin (AAT) to said subject.
  • a method of treating cytokine release syndrome or a cytokine storm in a subject undergoing chimeric antigen receptor- expressing T-cell (CAR T-cell) therapy comprises the step of administering a composition comprising Alpha- 1 -antitrypsin (AAT) to said subject.
  • a method of preventing cytokine release syndrome or a cytokine storm in a subject undergoing chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy comprises the step of administering a composition comprising Alpha- 1 -antitrypsin (AAT) to said subject.
  • a method of ameliorating cytokine release syndrome or a cytokine storm in a subject undergoing chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy comprises the step of administering a composition comprising Alpha- 1 -antitrypsin (AAT) to said subject.
  • a method of alleviating cytokine release syndrome or a cytokine storm in a subject undergoing chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy comprises the step of administering a composition comprising Alpha- 1 -antitrypsin (AAT) to said subject.
  • AAT Alpha- 1 -antitrypsin
  • a method of decreasing or inhibiting cytokine production in a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to cytokine release syndrome or cytokine storm comprising the step of administering a composition comprising Alpha- 1 -antitrypsin (AAT) to said subject.
  • AAT Alpha- 1 -antitrypsin
  • AAT is administered alone to control cytokine release.
  • both AAT and apoptotic cells or a composition thereof, or apoptotic cell supernatants or a composition thereof are administered to control cytokine release.
  • immune-modulating agents may encompass extracellular mediators, receptors, mediators of intracellular signaling pathways, regulators of translation and transcription, as well as immune cells.
  • an additional agent disclosed herein is an immune-modulatory agent known in the art.
  • use in the methods disclosed here of an immune-modulatory agent reduces the level of at least one cytokine.
  • use in the methods disclosed here of an immune-modulatory agent reduces or inhibits CRS or a cytokine storm.
  • an immune-modulatory agent comprises compounds that block, inhibit or reduce the release of cytokines or chemokines. In another embodiment, an immune-modulatory agent comprises compounds that block, inhibit or reduce the release of IL-21 or IL-23, or a combination thereof. In another embodiment, an immune-modulatory agent comprises an antiretroviral drug in the chemokine receptor-5 (CCR5) receptor antagonist class, for example maraviroc. In another embodiment, an immune-modulatory agent comprises an anti-DNAM-1 antibody. In another embodiment, an immune-modulatory agent comprises damage/pathogen- associated molecules (DAMPs/PAMPs) selected from the group comprising heparin sulfate, ATP, and uric acid, or any combination thereof.
  • DAMPs/PAMPs damage/pathogen- associated molecules
  • an immune-modulatory agent comprises a sialic acid binding Ig-like lectin (Siglecs).
  • an immune- modulatory agent comprises a cellular mediator of tolerance, for example regulatory CD4 + CD25 + T cells (Tregs) or invariant natural killer T cells (iNK T-cells).
  • an immune- modulatory agent comprises dendritic cells.
  • an immune-modulatory agent comprises monocytes.
  • an immune-modulatory agent comprises macrophages.
  • an immune-modulatory agent comprises JAK2 or JAK3 inhibitors selected from the group comprising ruxolitinib and tofacitinib.
  • an immune-modulatory agent comprises an inhibitor of spleen tyrosine kinase (Syk), for example fostamatinib.
  • an immune-modulatory agent comprises histone deacetylase inhibitor vorinostat acetylated STAT3.
  • an immune-modulatory agent comprises neddylation inhibitors, for example MLN4924.
  • an immune- modulatory agent comprises an miR-142 antagonist.
  • an immune- modulatory agent comprises a chemical analogue of cytidine, for example Azacitidine.
  • an immune-modulatory agent comprises an inhibitor of histone deacetylase, for example Vorinostat.
  • an immune-modulatory agent comprises an inhibitor of histone methylation.
  • Tellurium is a trace element found in the human body.
  • Various tellurium compounds have immune-modulating properties, and have been shown to have beneficial effects in diverse preclinical and clinical studies.
  • a particularly effective family of tellurium-containing compounds is disclosed for example, in U.S. Patent Nos. 4,752,614; 4,761,490; 4,764,461 and 4,929,739.
  • the immune-modulating properties of this family of tellurium-containing compounds is described, for example, in U.S. Patent Nos. 4,962,207, 5,093,135, 5,102,908 and 5,213,899, which are all incorporated by reference as if fully set forth herein.
  • AS 101 ammonium trichloro(dioxyethylene-0,0')tellurate, which is also referred to herein and in the art as AS 101.
  • AS 101 as a representative example of the family of tellurium-containing compound discussed hereinabove, exhibits antiviral (Nat. Immun. Cell Growth Regul. 7(3):163-8, 1988; AIDS Res Hum Retroviruses. 8(5):613-23, 1992), and tumoricidal activity (Nature 330(6144):173-6, 1987; J. Clin. Oncol. 13(9):2342-53, 1995; J. Immunol. 161(7):3536-42, 1998). Further, AS101 is characterized by low toxicity.
  • a composition comprising tellurium-containing immune-modulator compounds may be used in methods disclosed herein, where the tellurium-based compound stimulates the innate and acquired arm of the immune response.
  • AS101 is a potent activator of interferon (IFN) in mice (J. Natl. Cancer Inst. 88(18):1276-84, 1996) and humans (Nat. Immun. Cell Growth Regul. 9(3):182-90, 1990; Immunology 70(4):473-7, 1990; J. Natl. Cancer Inst. 88(18):1276-84, 1996.)
  • IFN interferon
  • tellurium-based compounds induce the secretion of a spectrum of cytokines, such as IL-la, IL-6 and TNF-a.
  • a tellurium-based compound comprises a tellurium-based compound known in the art to have immune-modulating properties.
  • a tellurium-based compound comprises ammonium trichloro(dioxyethylene-0,0')tellurate.
  • a tellurium-based compound inhibits the secretion of at least one cytokine.
  • a tellurium-based compound reduces the secretion of at least one cytokine.
  • a tellurium-based compound inhibits or reduces a cytokine release syndrome (CRS) of a cytokine storm.
  • CRS cytokine release syndrome
  • compositions comprising chimeric antigen receptor-expressing T-cells (CAR T-cells) and a tellurium-based compound.
  • CAR T-cells and Tellurium-based compound are in separate compositions.
  • AAT comprises a full length AAT or a functional fragment thereof.
  • AA comprises an analogue of a full length AAT or a functional fragment thereof
  • disclosed herein is a method of treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject comprising the step of administering chimeric antigen receptor-expressing T-cells (CAR T-cells) and a composition comprising a Tellurium-based compound to said subject.
  • CAR T-cells chimeric antigen receptor-expressing T-cells
  • a composition comprising a Tellurium-based compound
  • a method of inhibiting or reducing the incidence of cytokine release syndrome or cytokine storm in a subject undergoing chimeric antigen receptor- expressing T-cell (CAR T-cell) therapy comprising the step of administering a composition comprising a Tellurium-based compound to said subject.
  • a method of treating cytokine release syndrome or a cytokine storm in a subject undergoing chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy comprises the step of administering a composition comprising a Tellurium-based compound to said subject.
  • a method of preventing cytokine release syndrome or a cytokine storm in a subject undergoing chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy comprises the step of administering a composition comprising a Tellurium-based compound to said subject.
  • a method of ameliorating cytokine release syndrome or a cytokine storm in a subject undergoing chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy comprises the step of administering a composition comprising a Tellurium-based compound to said subject.
  • a method of alleviating cytokine release syndrome or a cytokine storm in a subject undergoing chimeric antigen receptor-expressing T-cell (CAR T-cell) therapy comprises the step of administering a composition comprising a Tellurium-based compound to said subject.
  • a method of decreasing or inhibiting cytokine production in a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to cytokine release syndrome or cytokine storm comprising the step of administering a composition comprising a Tellurium-based compound to said subject.
  • a tellurium-based compound is administered alone to control cytokine release.
  • both a tellurium-based compound and apoptotic cells or a composition thereof, or apoptotic cell supernatants or a composition thereof are administered to control cytokine release.
  • genetic modification of T-cells, dendritic cells, and/or apoptotic cells may be accomplished using RNA, DNA, recombinant viruses, or a combination thereof.
  • vectors derived from gamma retroviruses or lentiviruses are used in the compositions and methods as disclosed herein.
  • these vectors can integrate into the host genome, with potentially permanent expression of the transgene and have low intrinsic immunogenicity.
  • another vector that integrates into the host genome and/or has low intrinsic immunogenicity may be used in the compositions and methods as disclosed herein.
  • the non- viral- vector-mediated sleeping beauty transposon system is used to insert the CAR and other genes into the T-cell.
  • "suicide genes" are integrated into the T-cells, in which expression of a pro-apoptotic gene is under the control of an inducible promoter responsive to a systemically delivered drug.
  • genetic modification may be transient.
  • genetic modification may utilize messenger RNA (mRNA).
  • mRNA messenger RNA
  • large numbers of cells may be infused on multiple occasions in transiently engineered T-cells, such as mRNA-transfected T-cells.
  • RNA-based electroporation of lymphocytes using in vitro- transcribed mRNA mediates transient expression of proteins for approximately one week and obviates the risk of integrating viral vectors.
  • mRNA-transduced dendritic cells or mRNA-electroporated T and NK lymphocytes are examples of cells that are transient expression of proteins for approximately one week.
  • the genetic modification of the compositions and in the methods as disclosed herein may be any method that is known in the art.
  • apoptotic cells (Apocells) for use in compositions and methods as disclosed herein are as described in WO 2014/087408, which is incorporated by reference herein in its entirety.
  • apoptotic cells for use in compositions and methods as disclosed herein are produced in any way that is known in the art.
  • apoptotic cells for use in compositions and methods disclosed herein are autologous with a subject undergoing therapy.
  • apoptotic cells for use in compositions and methods disclosed herein are allogeneic with a subject undergoing therapy.
  • a composition comprising apoptotic cells comprises apoptotic cells as disclosed herein or as is known in art.
  • apoptotic cells comprise a cell preparation comprising mononuclear- enriched cells, wherein the preparation comprises at least 85% mononuclear cells, wherein at least 40% of the cells in the preparation are in an early-apoptotic state, wherein at least 85% of the cells in the preparation are viable cells and wherein the preparation comprises no more than 15% CD15 hlgh expressing cells.
  • the term "early-apoptotic state” may encompass cells that show early signs of apoptosis without late signs of apoptosis. Examples of early signs of apoptosis in cells include exposure of phosphatidylserine (PS) and the loss of mitochondrial membrane potential. Examples of late events include propidium iodide (PI) admission into the cell and the final DNA cutting.
  • PS phosphatidylserine
  • PI propidium iodide
  • cells that are stained by both Annexin-V FITC and PI are considered to be "late apoptotic cells”.
  • cells that do not stain for either Annexin-V or PI are considered non- apoptotic viable cells.
  • apoptotic cells comprise cells in an early apoptotic state. In another embodiment, apoptotic cells comprise cells wherein at least 90% of said cells are in an early apoptotic state. In another embodiment, apoptotic cells comprise cells wherein at least 80% of said cells are in an early apoptotic state. In another embodiment, apoptotic cells comprise cells wherein at least 70% of said cells are in an early apoptotic state. In another embodiment, apoptotic cells comprise cells wherein at least 60% of said cells are in an early apoptotic state. In another embodiment, apoptotic cells comprise cells wherein at least 50% of said cells are in an early apoptotic state.
  • early apoptotic cells are stable. In another embodiment, early apoptotic cells are stable for at least 24 hours. In another embodiment, early apoptotic cells are stable for 24 hours. In another embodiment, early apoptotic cells are stable for more than 24 hours. In another embodiment, early apoptotic cells are stable for at least 36 hours. In another embodiment, early apoptotic cells are stable for 48 hours. In another embodiment, early apoptotic cells are stable for at least 36 hours. In another embodiment, early apoptotic cells are stable for more than 36 hours. In another embodiment, early apoptotic cells are stable for at least 48 hours. In another embodiment, early apoptotic cells are stable for 48 hours.
  • early apoptotic cells are stable for at least 48 hours. In another embodiment, early apoptotic cells are stable for more than 48 hours. In another embodiment, early apoptotic cells are stable for at least 72 hours. In another embodiment, early apoptotic cells are stable for 72 hours. In another embodiment, early apoptotic cells are stable for at least 72 hours. In another embodiment, early apoptotic cells are stable for more than 72 hours.
  • stable encompasses apoptotic cells that remain PS-positive (Phosphatidylserine-positive) with only a very small percent of Pi-positive (Propidium iodide-positive).
  • Pi-positive cells provide an indication of membrane stability wherein a Pi-positive cells permits admission into the cells, showing that the membrane is less stable.
  • stable early apoptotic cells remain in early apoptosis for at least 24 hours, for at least 36 hours, for at least 48 hours, or for at least 72 hours.
  • stable early apoptotic cells remain in early apoptosis for 24 hours, for 36 hours, for 48 hours, or for 72 hours.
  • stable early apoptotic cells remain in early apoptosis for more than 24 hours, for more than 36 hours, for more than 48 hours, or for more than 72 hours. In another embodiment, stable early apoptotic cells maintain their state for an extended time period. In one embodiment, the composition comprising apoptotic cells further comprises an anti-coagulant.
  • the anti-coagulant is selected from the group consisting of: heparin, acid citrate dextrose (ACD) Formula A and a combination thereof.
  • the composition further comprises methylprednisolone.
  • the concentration of methylprednisolone does not exceed 30 ⁇ g/ml. In one embodiment, about 140 X 10 6 - 210 X 10 6 apoptotic cells are administered.
  • the apoptotic cells are used at a high dose. In one embodiment, the apoptotic cells are used at a high concentration. In one embodiment, human apoptotic polymorphonuclear neutrophils (PMNs) are used. In one embodiment, a group of cells, of which 50% are apoptotic cells, are used. In one embodiment, apoptotic cells are verified by May-Giemsa- stained cytopreps. In one embodiment, viability of cells are assessed by trypan blue exclusion. In one embodiment, the apoptotic and necrotic status of the cells are confirmed by annexin V/propidium iodide staining with detection by FACS.
  • PMNs human apoptotic polymorphonuclear neutrophils
  • apoptotic cells disclosed herein comprise no necrotic cells. In some embodiments, apoptotic cells disclosed herein comprise less than 1% necrotic cells. In some embodiments, apoptotic cells disclosed herein comprise less than 2% necrotic cells. In some embodiments, apoptotic cells disclosed herein comprise less than 3% necrotic cells. In some embodiments, apoptotic cells disclosed herein comprise less than 4% necrotic cells. In some embodiments, apoptotic cells disclosed herein comprise less than 5% necrotic cells.
  • a dose of lOxlO 6 apoptotic cells is administered.
  • a dose of lOxlO 7 apoptotic cells is administered.
  • 10x10 apoptotic cells is administered. In another embodiment, a dose of 10x10 apoptotic cells is administered. In another embodiment, a dose of lOxlO 10 apoptotic cells is administered. In another embodiment, a dose of lOxlO 11 apoptotic cells is administered. In another embodiment, a dose of lOxlO 12 apoptotic cells is administered. In another embodiment, a dose of 10x10 s apoptotic cells is administered. In another embodiment, a dose of lOxlO 4 apoptotic cells is administered. In another embodiment, a dose of 10x10 apoptotic cells is administered. In another embodiment, a dose of lOxlO 2 apoptotic cells is administered.
  • a high dose of apoptotic cells is administered.
  • a dose of 35xl0 6 apoptotic cells is administered.
  • a dose of 210xl0 6 apoptotic cells is administered.
  • a dose of 70x10 6 apoptotic cells is administered.
  • a dose of 140xl0 6 apoptotic cells is administered.
  • a dose of 35-210xl0 6 apoptotic cells is administered.
  • obtaining a mononuclear-enriched cell composition according to the production method disclosed herein is effected by leukapheresis.
  • leukapheresis may encompass an apheresis procedure in which leukocytes are separated from the blood of a donor.
  • the blood of a donor undergoes leukapheresis and thus a mononuclear-enriched cell composition is obtained according to the production method disclosed herein.
  • the use of at least one anticoagulant during leukapheresis is required, as is known in the art, in order to prevent clotting of the collected cells.
  • the leukapheresis procedure is configured to allow collection of mononuclear-enriched cell composition according to the production method disclosed herein.
  • cell collections obtained by leukapheresis comprise at least 65%. In other embodiments, at least 70%, or at least 80% mononuclear cells. Each possibility represents a separate embodiment as disclosed herein.
  • blood plasma from the cell-donor is collected in parallel to obtaining of the mononuclear-enriched cell composition according to the production method disclosed herein.
  • about 300-600ml of blood plasma from the cell-donor are collected in parallel to obtaining the mononuclear-enriched cell composition according to the production method disclosed herein.
  • blood plasma collected in parallel to obtaining the mononuclear-enriched cell composition according to the production method disclosed herein is used as part of the freezing and/or incubation medium.
  • Each possibility represents a separate embodiment as disclosed herein. Additional detailed methods of obtaining an enriched population of apoptotic cells for use in the compositions and methods as disclosed herein may be found in WO 2014/087408, which is incorporated herein by reference in its entirety.
  • the initial mononuclear- enriched cell preparation comprises at least 65% mononuclear cells, at least 70%, or at least 80% mononuclear cells
  • the final pharmaceutical composition disclosed herein, following the production method disclosed herein comprises at least 85%. In another embodiment, at least 90%, or at least 95% mononuclear cells.
  • Each possibility represents a separate embodiment as disclosed herein.
  • the apoptotic cells may be administered by any method known in the art including, but not limited to, intravenous, subcutaneous, intranodal, intratumoral, intrathecal, intrapleural, intraperitoneal and directly to the thymus.
  • the apoptotic cells are allogeneic. In one embodiment the apoptotic cells are from pooled third party donors. In one embodiment, the methods as disclosed herein comprise an additional step that is useful in overcoming rejection of allogeneic donor cells, including one or more steps described in U.S. Patent Application 20130156794, which is incorporated herein by reference in its entirety. In one embodiment, the methods comprise the step of full or partial lymphodepletion prior to administration of the apoptotic cells, which in one embodiment, are allogeneic apoptotic cells.
  • the lymphodepletion is adjusted so that it delays the host versus graft reaction for a period sufficient to allow the allogeneic apoptotic cells to control cytokine release.
  • the methods comprise the step of administering agents that delay egression of the allogeneic apoptotic T-cells from lymph nodes, such as 2-amino-2-[2-(4-octylphenyl)ethyl]propane-l,3-diol (FTY720), 5-[4-phenyl-5- (trifluoromemyl)thiophen-2-yl]-3-[3-(trifiuoromethyl)pheny- l]l,2,4-oxadiazole (SEW2871), 3-(2-(- hexylphenylamino)-2-oxoethylamino)propanoic acid (W123), 2-ammonio-4-(2-chloro-4-(3- phenoxyphenylthio)
  • the methods comprise the step of irradiating apoptotic cells derived from WBCs from a donor prior to administration to a recipient.
  • cells are irradiated in a way that will avoid proliferation and/or activation of residual viable cells within the apoptotic cell population.
  • the irradiated apoptotic cells preserve all their early apoptotic-, immune modulation-, stability-properties.
  • the irradiation step uses UV radiation.
  • the radiation step uses gamma radiation.
  • the apoptotic cells comprise a decreased percent of living non-apoptotic cells, comprise a preparation having a suppressed cellular activation of any living non-apoptotic cells present within the apoptotic cell preparation, or comprise a preparation having reduced proliferation of any living non-apoptotic cells present within the apoptotic cell preparation, or any combination thereof.
  • a pooled mononuclear apoptotic cell preparation comprising mononuclear cells in an early apoptotic state, wherein said pooled mononuclear apoptotic cells comprise a decreased percent of living non-apoptotic cells, a preparation having a suppressed cellular activation of any living non-apoptotic cells, or a preparation having reduced proliferation of any living non-apoptotic cells, or any combination thereof.
  • the pooled mononuclear apoptotic cells have been irradiated.
  • disclosed herein is a pooled mononuclear apoptotic cell preparation that in some embodiments, originates from the white blood cell fraction (WBC) obtained from donated blood.
  • WBC white blood cell fraction
  • the apoptotic cell preparation is irradiated.
  • said irradiation comprises gamma irradiation or UV irradiation.
  • the irradiated preparation has a reduced number of non-apoptotic cells compared with a non- irradiated apoptotic cell preparation.
  • the irradiated preparation has a reduced number of proliferating cells compared with a non-irradiated apoptotic cell preparation.
  • the irradiated preparation has a reduced number of potentially immunologically active cells compared with a non-irradiated apoptotic cell population.
  • pooled blood comprises 3rd party blood not matched between donor and recipient.
  • pooled may encompass blood collected from multiple donors, prepared and possibly stored for later use. This combined pool of blood may then be processed to produce a pooled mononuclear apoptotic cell preparation.
  • a pooled mononuclear apoptotic cell preparation ensures that a readily available supply of mononuclear apoptotic cells is available.
  • cells are pooled just prior to the incubation step wherein apoptosis is induced.
  • cells are pooled following the incubation step at the step of resuspension.
  • cells are pooled just prior to an irradiation step.
  • cells are pooled following an irradiation step.
  • cells are pooled at any step in the methods of preparation.
  • a pooled apoptotic cell preparation is derived from cells present in between about 2 and 25 units of blood.
  • said pooled apoptotic cell preparation is comprised of cells present in between about 2-5, 2-10, 2-15, 2-20, 5-10, 5-15, 5-20, 5- 25, 10-15, 10-20, 10-25, 6-13, or 6-25 units of blood.
  • said pooled apoptotic cell preparation is comprised of cells present in about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24 or 25 units of blood. The number of units of blood needed is also dependent upon the efficiency of WBC recovery from blood.
  • each unit is a bag of blood.
  • a pooled apoptotic cell preparation is comprised of cells present in at least 25 units of blood, at least 50 units of blood, or at least 100 units of blood. Each possibility represents a separate embodiment as disclosed herein.
  • the units of blood comprise white blood cell (WBC) fractions from blood donations.
  • the donations may be from a blood center or blood bank.
  • the donations may be from donors in a hospital gathered at the time of preparation of the pooled apoptotic cell preparation.
  • units of blood comprising WBCs from multiple donors are saved and maintained in an independent blood bank created for the purpose of compositions and methods thereof as disclosed herein.
  • a blood bank developed for the purpose of compositions and methods thereof as disclosed herein is able to supply units of blood comprising WBC from multiple donors and comprises a leukapheresis unit.
  • the units of pooled WBCs are not restricted by HLA matching. Therefore, the resultant pooled apoptotic cell preparation comprises cell populations not restricted by HLA matching. Accordingly, in certain embodiments a pooled mononuclear apoptotic cell preparation comprises allogeneic cells.
  • An advantage of a pooled mononuclear apoptotic cell preparation that is derived from pooled WBCs not restricted by HLA matching, is a readily available source of WBCs and reduced costs of obtaining WBCs.
  • pooled blood comprises blood from multiple donors independent of HLA matching.
  • pooled blood comprises blood from multiple donors wherein HLA matching with the recipient has been taken into consideration. For example, wherein 1 HLA allele, 2 HLA alleles, 3 HLA alleles, 4 HLA alleles, 5 HLA alleles, 6 HLA alleles, or 7 HLA alleles have been matched between donors and recipient.
  • multiple donors are partially matched, for example some of the donors have been HLA matched wherein 1 HLA allele, 2 HLA alleles, 3 HLA alleles, 4 HLA alleles, 5 HLA alleles, 6 HLA alleles, or 7 HLA alleles have been matched between some of the donors and recipient.
  • Each possibility comprises an embodiment as disclosed herein.
  • some viable non-apoptotic cells may remain following the induction of apoptosis step described below.
  • the presence of these viable non- apoptotic cells is, in one embodiment, observed prior to an irradiation step.
  • These viable non- apoptotic cells may be able to proliferate or be activated.
  • the pooled mononuclear apoptotic cell preparation derived from multiple donors may be activated against the host, activated against one another, or both.
  • an irradiated cell preparation as disclosed herein has suppressed cellular activation and reduced proliferation compared with a non-irradiated cell preparation.
  • the irradiation comprises gamma irradiation or UV irradiation.
  • an irradiated cell preparation has a reduced number of non-apoptotic cells compared with a non-irradiated cell preparation.
  • the irradiation comprises about 15 Grey units (Gy). In another embodiment, the irradiation comprises about 20 Grey units (Gy). In another embodiment, the irradiation comprises about 25 Grey units (Gy). In another embodiment, the irradiation comprises about 30 Grey units (Gy).
  • the irradiation comprises about 35 Grey units (Gy). In another embodiment, the irradiation comprises about 40 Grey units (Gy). In another embodiment, the irradiation comprises about 45 Grey units (Gy). In another embodiment, the irradiation comprises about 50 Grey units (Gy). In another embodiment, the irradiation comprises about 55 Grey units (Gy). In another embodiment, the irradiation comprises about 60 Grey units (Gy). In another embodiment, the irradiation comprises about 65 Grey units (Gy). In another embodiment, the irradiation comprises up to 2500 Gy. In another embodiment, an irradiated pooled apoptotic cell preparation maintains the same or a similar apoptotic profile, stability and efficacy as a non-irradiated pooled apoptotic cell preparation.
  • a pooled mononuclear apoptotic cell preparation as disclosed herein is stable for up to 24 hours. In another embodiment, a pooled mononuclear apoptotic cell preparation is stable for at least 24 hours. In another embodiment, a pooled mononuclear apoptotic cell preparation is stable for more than 24 hours. In yet another embodiment, a pooled mononuclear apoptotic cell preparation as disclosed herein is stable for up to 36 hours. In still another embodiment, a pooled mononuclear apoptotic cell preparation is stable for at least 36 hours. In a further embodiment, a pooled mononuclear apoptotic cell preparation is stable for more than 36 hours.
  • a pooled mononuclear apoptotic cell preparation as disclosed herein is stable for up to 48 hours. In another embodiment, a pooled mononuclear apoptotic cell preparation is stable for at least 48 hours. In another embodiment, a pooled mononuclear apoptotic cell preparation is stable for more than 48 hours.
  • methods of producing the pooled cell preparation comprising an irradiation step preserves the early apoptotic, immune modulation, and stability properties observed in an apoptotic preparation derived from a single match donor wherein the cell preparation may not include an irradiation step.
  • a pooled mononuclear apoptotic cell preparation as disclosed herein does not elicit a graft versus host disease (GVHD) response.
  • GVHD graft versus host disease
  • Irradiation of the cell preparation is considered safe in the art. Irradiation procedures are currently performed on a routine basis to donated blood to prevent reactions to WBC.
  • the percent of apoptotic cells in a pooled mononuclear apoptotic cell preparation as disclosed herein is close to 100%, thereby reducing the fraction of living non- apoptotic cells in the cell preparation.
  • the percent of apoptotic cells is at least 40%.
  • the percent of apoptotic cells is at least 50%.
  • the percent of apoptotic cells is at least 60%.
  • the percent of apoptotic cells is at least 70%.
  • the percent of apoptotic cells is at least 80%.
  • the percent of apoptotic cells is at least 90%.
  • the percent of apoptotic cells is at least 99%. Accordingly, a cell preparation comprising a reduced or non-existent fraction of living non-apoptotic cells may in one embodiment provide a pooled mononuclear apoptotic cell preparation that does not elicit GVHD in a recipient. Each possibility represents an embodiment as disclosed herein.
  • the percentage of living non-apoptotic WBC is reduced by specifically removing the living cell population, for example by targeted precipitation.
  • the percent of living non-apoptotic cells may be reduced using magnetic beads that bind to phosphatidylserine.
  • the percent of living non-apoptotic cells may be reduced using magnetic beads that bind a marker on the cell surface of non-apoptotic cells but not apoptotic cells.
  • the apoptotic cells may be selected for further preparation using magnetic beads that bind to a marker on the cell surface of apoptotic cells but not non-apoptotic cells.
  • the percentage of living non-apoptotic WBC is reduced by the use of ultrasound.
  • the apoptotic cells are from pooled third party donors.
  • a pooled cell preparation comprises at least one cell type selected from the group consisting of: lymphocytes, monocytes and natural killer cells.
  • a pooled cell preparation comprises an enriched population of mononuclear cells.
  • a pooled mononuclear is a mononuclear enriched cell preparation comprises cell types selected from the group consisting of: lymphocytes, monocytes and natural killer cells.
  • the mononuclear enriched cell preparation comprises no more than 15%, alternatively no more than 10%, typically no more than 5% polymorphonuclear leukocytes, also known as granulocytes (i.e., neutrophils, basophils and eosinophils).
  • a pooled mononuclear cell preparation is devoid of granulocytes. Each possibility represents a separate embodiment as disclosed herein.
  • the pooled mononuclear enriched cell preparation comprises no more than 15%, alternatively no more than 10%, typically no more than 5% CD15 Mgh expressing cells. In one embodiment, a pooled apoptotic cell preparation comprises less than 15% CD 15 high expressing cells.
  • the pooled mononuclear enriched cell preparation disclosed herein comprises at least 80% mononuclear cells, at least 85% mononuclear cells, alternatively at least 90% mononuclear cells, or at least 95% mononuclear cells, wherein each possibility is a separate embodiment disclosed herein. According to some embodiments, the pooled mononuclear enriched cell preparation disclosed herein comprises at least 85% mononuclear cells.
  • any pooled cell preparation that has a final pooled percent of mononuclear cells of at least 80% is considered a pooled mononuclear enriched cell preparation as disclosed herein.
  • pooling cell preparations having increased polymorphonuclear cells (PMN) with cell preparations having high mononuclear cells with a resultant "pool" of at least 80% mononuclear cells comprises a preparation as disclosed herein.
  • mononuclear cells comprise lymphocytes and monocytes.
  • a pooled apoptotic cell preparation as disclosed herein comprises less than 5% polymorphonuclear leukocytes.
  • the apoptotic cells are T-cells. In another embodiment, the apoptotic cells are derived from the same pooled third party donor T-cells as the CAR T-cells. In another embodiment, the apoptotic cells are derived from the CAR T-cell population.
  • the apoptotic cells reduce production of cytokines associated with the cytokine storm including but not limited to IL-6, and interferon-gamma (IFN- ⁇ ), alone or in combination, while the effectiveness of CAR T-cell therapy was maintained (Example 2).
  • the apoptotic cells affect cytokine expression levels in macrophages.
  • the apoptotic cells reduce cytokine expression levels in macrophages.
  • the apoptotic cells suppress cytokine expression levels in macrophages.
  • the apoptotic cells inhibit cytokine expression levels in macrophages.
  • the apoptotic cells maintain IFN- ⁇ levels to match or nearly match levels present prior to CAR -T cell administration.
  • apoptotic cells affect cytokine expression levels in macrophages but do not affect cytokine expression levels in the CAR T-cells.
  • the apoptotic cells affect cytokine expression levels in DCs, but do not affect cytokine expression levels in the CAR T-cells. It was therefore unexpected that apoptotic cells would be useful in maintaining the effectiveness CAR T-cell therapy.
  • the effect of apoptotic cells on cytokine expression levels in macrophages, DCs, or a combination thereof results in reduction of CRS.
  • the effect of apoptotic cells on cytokine expression levels in macrophages, DCs, or a combination thereof results in reduction of severe CRS.
  • the effect of apoptotic cells on cytokine expression levels in macrophages, DCs, or a combination thereof results in suppression of CRS.
  • the effect of apoptotic cells on cytokine expression levels in macrophages, DCs, or a combination thereof results in suppression of severe CRS.
  • the effect of apoptotic cells on cytokine expression levels in macrophages, DCs, or a combination thereof results in inhibition of CRS.
  • the effect of apoptotic cells on cytokine expression levels in macrophages, DCs, or a combination thereof results in inhibition of severe CRS.
  • the effect of apoptotic cells on cytokine expression levels in macrophages, DCs, or a combination thereof results in prevention of CRS.
  • the effect of apoptotic cells on cytokine expression levels in macrophages, DCs, or a combination thereof results in prevention of severe CRS.
  • the apoptotic cells trigger death of T-cells, but not via changes in cytokine expression levels.
  • apoptotic cells antagonize the priming of macrophages and dendritic cells to secrete cytokines that would otherwise amplify the cytokine storm.
  • apoptotic cells increase Tregs which suppress the inflammatory response and/or prevent excess release of cytokines.
  • administration of apoptotic cells inhibits one or more pro-inflammatory cytokines.
  • the pro-inflammatory cytokine comprises IL-lbeta, IL-6, TNF- alpha, or IFN-gamma, or any combination thereof.
  • administration of apoptotic cells promotes the secretion of one or more anti-inflammatory cytokines.
  • the anti-inflammatory cytokine comprises TGF-beta, IL10, or PGE2, or any combination thereof.
  • administration of apoptotic cells inhibits dendritic cell maturation following exposure to TLR ligands.
  • administration of apoptotic cells creates potentially tolerogenic dendritic cells, which in one embodiment, are capable of migration, and in one embodiment, the migration is due to CCR7.
  • administration of apoptotic cells elicits various signaling events which in one embodiment is TAM receptor signaling (Tyro3, Axl and Mer) which in one embodiment, inhibits inflammation in antigen-presenting cells.
  • Tyro-3, Axl, and Mer constitute the TAM family of receptor tyrosine kinases (RTKs) characterized by a conserved sequence within the kinase domain and adhesion moleculelike extracellular domains.
  • administration of apoptotic cells activates signaling through MerTK.
  • administration of apoptotic cells activates the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, which in one embodiment, negatively regulates NF- ⁇ .
  • administration of apoptotic cells negatively regulates the inflammasome which in one embodiment leads to inhibition of pro-inflammatory cytokine secretion, DC maturation, or a combination thereof.
  • administration of apoptotic cells upregulates expression of anti-inflammatory genes such as Nr4a, Thbsl, or a combination thereof.
  • administration of apoptotic cells induces a high level of AMP which in one embodiment, is accumulated in a Pannexinl -dependent manner.
  • administration of apoptotic cells suppresses inflammation
  • compositions for use in the methods and treatments as disclosed herein include an apoptotic cell supernatant as disclosed herein.
  • the apoptotic cell supernatant is obtained by a method comprising the steps of a) providing apoptotic cells, b) culturing the apoptotic cells of step a), and c) separating the supernatant from the cells.
  • apoptotic cells for use making an apoptotic cell supernatant as disclosed herein are autologous with a subject undergoing therapy.
  • apoptotic cells for use in making an apoptotic cell supernatant disclosed herein are allogeneic with a subject undergoing therapy.
  • the apoptotic cells from which the apoptotic cell supernatant is obtained may be cells chosen from any cell type of a subject, or any commercially available cell line, subjected to a method of inducing apoptosis known to the person skilled in the art.
  • the method of inducing apoptosis may be hypoxia, ozone, heat, radiation, chemicals, osmotic pressure, pH shift, X-ray irradiation, gamma- ray irradiation, UV irradiation, serum deprivation, corticoids or combinations thereof, or any other method described herein or known in the art.
  • the method of inducing apoptosis produces apoptotic cells in an early apoptotic state.
  • the apoptotic cells are leukocytes.
  • said apoptotic leukocytes are derived from peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • said leukocytes are from pooled third party donors.
  • said leukocytes are allogeneic.
  • the apoptotic cells are provided by selecting non-adherent leukocytes and submitting them to apoptosis induction, followed by a cell culture step in culture medium.
  • "Leukocytes" used to make the apoptotic cell-phagocyte supernatant may be derived from any lineage, or sub-lineage, of nucleated cells of the immune system and/or hematopoietic system, including but not limited to dendritic cells, macrophages, masT-cells, basophils, hematopoietic stem cells, bone marrow cells, natural killer cells, and the like.
  • the leukocytes may be derived or obtained in any of various suitable ways, from any of various suitable anatomical compartments, according to any of various commonly practiced methods, depending on the application and purpose, desired leukocyte lineage, etc.
  • the source leukocytes are primary leukocytes.
  • the source leukocytes are primary peripheral blood leukocytes.
  • Primary lymphocytes and monocytes may be conveniently derived from peripheral blood.
  • Peripheral blood leukocytes include 70-95 percent lymphocytes, and 5-25 percent monocytes.
  • Source lymphocytes and/or monocytes can be achieved, for example, by harvesting blood in the presence of an anticoagulant, such as heparin or citrate. The harvested blood is then centrifuged over a Ficoll cushion to isolate lymphocytes and monocytes at the gradient interface, and neutrophils and erythrocytes in the pellet.
  • an anticoagulant such as heparin or citrate. The harvested blood is then centrifuged over a Ficoll cushion to isolate lymphocytes and monocytes at the gradient interface, and neutrophils and erythrocytes in the pellet.
  • Leukocytes may be separated from each other via standard immunomagnetic selection or immunofiuorescent flow cytometry techniques according to their specific surface markers, or via centrifugal elutriation.
  • monocytes can be selected as the CD 14+ fraction
  • T- lymphocytes can be selected as CD3+ fraction
  • B-lymphocytes can be selected as the CD 19+ fraction
  • macrophages as the CD206+ fraction.
  • Lymphocytes and monocytes may be isolated from each other by subjecting these cells to substrate-adherent conditions, such as by static culture in a tissue culture-treated culturing recipient, which results in selective adherence of the monocytes, but not of the lymphocytes, to the cell- adherent substrate.
  • Leukocytes may also be obtained from peripheral blood mononuclear cells (PBMCs), which may be isolated as described herein.
  • PBMCs peripheral blood mononuclear cells
  • Suitable leukocyte cell lines may be obtained from commercial suppliers, such as the American Tissue Type Collection (ATCC). It will be evident to the person skilled in the art that source leukocytes should not be obtained via a technique which will significantly interfere with their capacity to produce the apoptotic leukocytes.
  • ATCC American Tissue Type Collection
  • the apoptotic cells comprise a cell preparation comprising mononuclear- enriched cells, wherein the preparation comprises at least 85% mononuclear cells, wherein at least 40% of the cells in the preparation are in an early-apoptotic state, wherein at least 85% of the cells in the preparation are viable cells and wherein the preparation comprises no more than 15% CD15 hlgh expressing cells.
  • the apoptotic cells may be apoptotic lymphocytes.
  • Apoptosis of lymphocytes such as primary lymphocytes, may be induced by treating the primary lymphocytes with serum deprivation, a corticosteroid, or irradiation.
  • inducing apoptosis of primary lymphocytes via treatment with a corticosteroid is effected by treating the primary lymphocytes with dexamethasone.
  • dexamethasone at a concentration of about 1 micromolar.
  • inducing apoptosis of primary lymphocytes via irradiation is effected by treating the primary lymphocytes with gamma- irradiation.
  • with a dosage of about 66 rad Such treatment results in the generation of apoptotic lymphocytes suitable for the co-culture step with phagocytes.
  • apoptotic cells may be apoptotic monocytes, such as primary monocytes.
  • apoptotic monocytes such as primary monocytes.
  • the monocytes are subjected to in vitro conditions of substrate/surface-adherence under conditions of serum deprivation. Such treatment results in the generation of non-pro-inflammatory apoptotic monocytes suitable for the co-culture step with phagocytes.
  • the apoptotic cells may be any apoptotic cells described herein, including allogeneic apoptotic cells, third party apoptotic cells, and pools of apoptotic cells.
  • the apoptotic cell supernatant may be obtained through the co-culture of apoptotic cells with other cells.
  • the apoptotic cell supernatant is an apoptotic cell supernatant obtained by a method comprising the steps of a) providing apoptotic cells, b) providing other cells, c) optionally washing the cells from step a) and b), d) co-culturing the cells of step a) and b), and optionally e) separating the supernatant from the cells.
  • the other cells co-cultured with the apoptotic cells are white blood cells.
  • the apoptotic cell supernatant is an apoptotic cell-white blood cell supernatant obtained by a method comprising the steps of a) providing apoptotic cells, b) providing white blood cells, c) optionally washing the cells from step a) and b), d) co-culturing the cells of step a) and b), and optionally e) separating the supernatant from the cells.
  • the white blood cells may be phagocytes, such as macrophages, monocytes or dendritic cells.
  • the white blood cells may be B cells, T-cells, or natural killer (NK cells).
  • compositions for use in the methods and treatments as disclosed herein include apoptotic cell-phagocyte supernatants as described in WO 2014/106666, which is incorporated by reference herein in its entirety.
  • apoptotic cell-phagocyte supernatants for use in compositions and methods as disclosed herein are produced in any way that is known in the art.
  • the apoptotic supernatant comprises an apoptotic cell-phagocyte supernatant that is obtained from a co-culture of phagocytes with apoptotic cells
  • the apoptotic cell-phagocyte supernatant is obtained by a method comprising the steps of a) providing phagocytes, b) providing apoptotic cells, c) optionally washing the cells from step a) and b), d) co-culturing the cells of step a) and b), and optionally e) separating the supernatant from the cells.
  • an apoptotic supernatant comprises a supernatant produced by phagocytic cells that ingest the apoptotic cells.
  • phagocytes denotes cells that protect the body by ingesting (phagocytosing) harmful foreign particles, bacteria, and dead or dying cells.
  • Phagocytes include for example cells called neutrophils, monocytes, macrophages, dendritic cells, and mast T-cells, preferentially dendritic cells and monocytes/macrophages.
  • the phagocytes may be dendritic cells (CD4+ HLA- DR+ Lineage- BDCA1 /BDCA3+), macrophages (CD14+ CD206+ HLA-DR+), or derived from monocytes (CD14+). Techniques to distinguish these different phagocytes are known to the person skilled in the art.
  • monocytes are obtained by a plastic adherence step. Said monocytes can be distinguished from B and T-cells with the marker CD14+, whereas unwanted B cells express CD19+ and T-cells CD3+. After Macrophage Colony Stimulating Factor (M-CSF) induced maturation the obtained macrophages are in one embodiment, positive for the markers CD 14+, CD206+, HLA-DR+.
  • M-CSF Macrophage Colony Stimulating Factor
  • said phagocytes are derived from peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • Phagocytes may be provided by any method known in the art for obtaining phagocytes.
  • phagocytes such as macrophages or dendritic cells can be directly isolated from a subject or be derived from precursor cells by a maturation step.
  • macrophages may be directly isolated from the peritoneum cavity of a subject and cultured in complete RRPMI medium. Macrophages can also be isolated from the spleen.
  • Phagocytes are also obtainable from peripheral blood monocytes.
  • monocytes when cultured differentiate into monocyte-derived macrophages upon addition of, without limitation to, macrophage colony stimulating factor (M-CSF) to the cell culture media.
  • M-CSF macrophage colony stimulating factor
  • phagocytes may be derived from peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • Non-adherent T-cells are removed by a plastic adherence step, and adherent T-cells cultured in complete RPMI milieu supplemented with recombinant human M-CSF. After the culture period, monocyte-derived macrophages are obtained.
  • Phagocytes can be selected by a cell- adherence step.
  • Said "cell adherence step” means that phagocytes or cells which can mature into phagocytes are selected via culturing conditions allowing the adhesion of the cultured cells to a surface, a cell adherent surface (e.g. a tissue culture dish, a matrix, a sac or bag with the appropriate type of nylon or plastic).
  • a cell adherent surface e.g. a tissue culture dish, a matrix, a sac or bag with the appropriate type of nylon or plastic.
  • Cell adherent surfaces may encompass hydrophilic and negatively charged, and may be obtained in any of various ways known in the art, In another embodiment by modifying a polystyrene surface using, for example, corona discharge, or gas-plasma.
  • B cells, T-cells and NK cells may be provided by any method known in the art for obtaining such cells.
  • B cells, T-cells or NK cells can be directly isolated from a subject or be derived from precursor cells by a maturation step.
  • the B, T or NK cells can be from a B, T or NK cell line.
  • One of ordinary skill in the art will possess the necessary expertise to establish, purchase, or otherwise obtain suitable established B cells, T-cells and NK cell lines. Suitable cell lines may be obtained from commercial suppliers, such as the American Tissue Type Collection (ATCC).
  • ATCC American Tissue Type Collection
  • said apoptotic cells and said white blood cells are cultured individually prior to the co-culture step d).
  • the cell maturation of phagocytes takes place during cell culture, for example due to addition of maturation factors to the media.
  • said maturation factor is M-CSF, which may be used for example to obtain monocyte-derived macrophages.
  • the culture step used for maturation or selection of phagocytes might take several hours to several days.
  • said pre-mature phagocytes are cultured for 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58 hours in an appropriate culture medium.
  • the culture medium for phagocytes is known to the person skilled in the art and can be for example, without limitation, RPMI, DMEM, X-vivo and Ultraculture milieus.
  • co-culture of apoptotic cells and phagocytes takes place in a physiological solution.
  • the cells Prior to this "co-culture", the cells may be submitted to a washing step.
  • the white blood cells e.g. the phagocytes
  • the apoptotic cells are washed before the co-culture step.
  • the cells are washed with PBS.
  • the white blood cells e.g. the phagocytes such as macrophages, monocytes, or phagocytes, or the B, T or NK cells
  • the apoptotic cells may be mixed in a ratio of 10:1, 9:1; 8:1 , 7:1, 6:1, 5:1 , 4:1, 3:1 , 2:1, or 1 :1, or in a ratio of (white blood cells : apoptotic cells) 1 :2, 1:3, 1 :4, 1 :5, 1:6, 1 :7, 1 :8, 1:9, or 1 :10.
  • the ratio of white blood cells to apoptotic cells is 1 :5.
  • the co-culture of the cells might be for several hours to several days.
  • said apoptotic cells are cultured for 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52 hours.
  • a person skilled in the art can evaluate the optimal time for co- culture by measuring the presence of anti-inflammatory compounds, the viable amount of white blood cells and the amount of apoptotic cells which have not been eliminated so far.
  • the culture of apoptotic cells takes place in culture medium and/or in a physiological solution compatible with administration e.g. injection to a subject.
  • white blood cells e.g. phagocytes such as macrophages, monocytes, or phagocytes, or the B, T or NK cells
  • a "physiological solution” may encompass a solution which does not lead to the death of white blood cells within the culture time. In some embodiments, the physiological solution does not lead to death over 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52 hours. In other embodiment, 48 hours, or 30 hours.
  • the white blood cells e.g. phagocytes such as macrophages, monocytes, or phagocytes, or the B, T or NK cells
  • the apoptotic cells are incubated in the physiological solution for at least 30 min. This time of culture allows phagocytosis initiation and secretion of cytokines and other beneficial substances.
  • such a physiological solution does not inhibit apoptotic leukocyte elimination by leukocyte-derived macrophages.
  • the supernatant is optionally separated from the cultured apoptotic cells or the co-cultured cells.
  • Techniques to separate the supernatant from the cells are known in the art.
  • the supernatant may be collected and/or filtered and/or centrifuged to eliminate cells and debris.
  • the supernatant may be centrifuged at 3000 rpm for 15 minutes at room temperature to separate it from the cells.
  • the supernatant may be "inactivated" prior to use, for example by irradiation. Therefore, the method for preparing the apoptotic cell supernatant may comprise an optional additional irradiation step f). Said "irradiation" step can be considered as a disinfection method that uses X-ray irradiation (25-45 Gy) at sufficiently rate to kill microorganisms, as routinely performed to inactivate blood products.
  • Irradiation of the supernatant is considered safe in the art. Irradiation procedures are currently performed on a routine basis to donated blood to prevent reactions to WBC.
  • the apoptotic cell supernatant is formulated into a pharmaceutical composition suitable for administration to a subject, as described in detail herein.
  • the final product is stored at +4°C. In another embodiment, the final product is for use in the next 48 hours.
  • the apoptotic cell supernatant such as an apoptotic cell-phagocyte supernatant, or pharmaceutical composition comprising the supernatant, may be lyophilized, for example for storage at -80 °C.
  • an apoptotic cell-phagocyte supernatant may be made using thymic cells as apoptotic cells.
  • thymic cells are irradiated (e.g. with a 35 X-Gray irradiation) and cultured in complete DMEM culture medium for, for example, 6 hours to allow apoptosis to occur.
  • macrophages are isolated from the peritoneum cavity, washed and cultured in complete RPMI (10% FBS, Peni- Strepto, EAA, Hepes, NaP and 2-MercaptoEthanol).
  • Macrophages and apoptotic cells are then washed and co-cultured for another 48 hour period in phenol-free X-vivo medium at a 1/5 macrophage/apoptotic cell ratio. Then, supernatant is collected, centrifuged to eliminate debris and may be frozen or lyophilized for conservation. Macrophage enrichment may be confirmed using positive staining for F4/80 by FACS. Apoptosis may be confirmed by FACS using positive staining for Annexin-V and 7AAD exclusion.
  • the apoptotic cell supernatant is enriched in TGF- ⁇ levels both in active and latent forms of TGF- ⁇ , compared to supernatants obtained from either macrophages or apoptotic cells cultured separately.
  • IL-10 levels are also increased compared to macrophages cultured alone and dramatically increased compared to apoptotic cells cultured alone.
  • inflammatory cytokines such as IL-6 are not detectable and IL-1 ⁇ and TNF are undetectable or at very low levels.
  • the apoptotic cell supernatant when compared to supernatants from macrophages cultured alone or from apoptotic cells cultured alone, has increased levels of IL-lra, TIMP-1, CXCL1/KC and CCL2/JE/MCP1, which might be implicated in a tolerogenic role of the supernatant to control inflammation, in addition to TGF- ⁇ and IL-10.
  • human apoptotic cell-phagocyte supernatant may be made from the co-culture of macrophages derived from peripheral blood mononuclear cells (PBMC) cultured with apoptotic PBMC.
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • non-adherent-cells are removed and rendered apoptotic using, for example, a 35 Gy dose of X-ray irradiation and cultured in complete RPMI milieu for 4 days (including cell wash after the first 48 hrs of culture), in order to allow apoptosis to occur.
  • non-adherent T-cells are cultured in complete RPMI milieu supplemented with 50 ⁇ g/mL of recombinant human M-CSF for 4 days including cell wash after the first 48 hrs.
  • monocyte-derived macrophages and apoptotic cells are washed and cultured together in X-vivo medium for again 48 hours at a one macrophage to 5 apoptotic cell ratio. Then supernatant from the latter culture is collected, centrifuged to eliminate cells and debris, and may be frozen or lyophilized for conservation and subsequent use.
  • human apoptotic cell-phagocyte supernatant may be obtained in 6 days from peripheral blood mononuclear cells (PBMC). Four days to obtain PBMC- derived macrophages using M-CSF addition in the culture, and 2 more days for the co-culture of PBMC-derived macrophages with apoptotic cells, corresponding to the nonadherent PBMC isolated at day 0.
  • PBMC peripheral blood mononuclear cells
  • a standardized human apoptotic cell- phagocyte supernatant may be obtained independently of the donor or the source of PBMC (cytapheresis or buffy coat).
  • the plastic-adherence step is sufficient to obtain a significant starting population of enriched monocytes (20 to 93% of CD14+ cells after adherence on plastic culture dish).
  • adherent cells demonstrate a very low presence of B and T-cells (1.0% of CD19+ B cells and 12.8% of CD3+ T-cells).
  • monocyte-derived macrophages After 4 days of culture of T-cells in the presence of M- CSF, the proportion of monocytes derived- macrophages is significantly increased from 0.1 % to 77.7% of CD14+CD206+HLA-DR+ macrophages. At that time, monocyte-derived macrophages may be co-cultured with apoptotic non-adherent PBMC (47.6% apoptotic as shown by annexin V staining and 7AAD exclusion) to produce the apoptotic cell-phagocyte supernatant during 48 hours.
  • apoptotic non-adherent PBMC 47.6% apoptotic as shown by annexin V staining and 7AAD exclusion
  • the collected apoptotic cell-phagocyte supernatant contains significantly more latent TGF than in the culture supernatant of monocyte-derived macrophages alone or monocyte-derived macrophages treated in inflammatory conditions (+ LPS), and only contains trace or low level of inflammatory cytokines such as IL- ⁇ or TNF.
  • the composition comprising the apoptotic cell supernatant further comprises an anti-coagulant.
  • the anti-coagulant is selected from the group consisting of: heparin, acid citrate dextrose (ACD) Formula A and a combination thereof.
  • an anti-coagulant is added during the process of manufacturing apoptotic cells.
  • the anti-coagulant added is selected from the group comprising ACD and heparin, or any combination thereof.
  • ACD is at a concentration of 1%.
  • ACD is at a concentration of 2%.
  • ACD is at a concentration of 3%.
  • ACD is at a concentration of 4%.
  • ACD is at a concentration of 5%.
  • ACD is at a concentration of 6%.
  • ACD is at a concentration of 7%.
  • ACD is at a concentration of 8%.
  • ACD is at a concentration of 9%.
  • ACD is at a concentration of 10%. In another embodiment, ACD is at a concentration of between about 1-10%. In another embodiment, ACD is at a concentration of between about 2-8 %. In another embodiment, ACD is at a concentration of between about 3-7%. In another embodiment, ACD is at a concentration of between about 1-5%. In another embodiment, ACD is at a concentration of between about 5-10%. In another embodiment, heparin is at a final concentration of 0.5 U/ml. In another embodiment, heparin is at a final concentration of about 0.1 U/ml- 1.0 U/ml. In another embodiment, heparin is at a final concentration of about 0.2 U/ml-0.9 U/ml.
  • heparin is at a final concentration of about 0.3 U/ml-0.7 U/ml. In another embodiment, heparin is at a final concentration of about 0. 1 U/ml-0.5 U/ml. In another embodiment, heparin is at a final concentration of about 0.5 U/ml- 1.0 U/ml. In another embodiment, heparin is at a final concentration of about 0.01 U/ml- 1.0 U/ml. In another embodiment, heparin is at a final concentration of 0.1 U/ml. In another embodiment, heparin is at a final concentration of 0.2 U/ml. In another embodiment, heparin is at a final concentration of 0.3 U/ml.
  • heparin is at a final concentration of 0.4 U/ml. In another embodiment, heparin is at a final concentration of 0.5 U/ml. In another embodiment, heparin is at a final concentration of 0.6 U/ml. In another embodiment, heparin is at a final concentration of 0.7 U/ml. In another embodiment, heparin is at a final concentration of 0.8 U/ml. In another embodiment, heparin is at a final concentration of 0.9 U/ml. In another embodiment, heparin is at a final concentration of 1.0 U/ml. In another embodiment, ACD is at a concentration of 5% and heparin is at a final concentration of 0.5 U/ml.
  • the composition comprising the apoptotic cell supernatant further comprises methylprednisolone.
  • the concentration of methylprednisolone does not exceed 30 ⁇ .
  • the composition may be used at a total dose or aliquot of apoptotic cell supernatant derived from the co-culture of about 14xl0 9 of CD45+ cells obtained by cytapheresis equivalent to about 200 million of cells per kilogram of body weight (for a 70 kg subject).
  • a total dose is administered as unit doses of supernatant derived from about 100 million cells per kilogram body weight, and/or is administered as unit doses at weekly intervals, In another embodiment both of which.
  • Suitable total doses according to this embodiment include total doses of supernatant derived from about 10 million to about 4 billion cells per kilogram body weight.
  • the supernatant is derived from about 40 million to about 1 billion cells per kilogram body weight. In yet another embodiment the supernatant is derived from about 80 million to about 500 million cells per kilogram body weight. In still another embodiment, the supernatant is derived from about 160 million to about 250 million cells per kilogram body weight. Suitable unit doses according to this embodiment include unit doses of supernatant derived from about 4 million to about 400 million cells per kilogram body weight. In another embodiment, the supernatant is derived from about 8 million to about 200 million cells per kilogram body weight. In another embodiment, the supernatant is derived from about 16 million to about 100 million cells per kilogram body weight. In yet another embodiment, the supernatant is derived from about 32 million to about 50 million cells per kilogram body weight.
  • a dose of apoptotic cell supernatant derived from the co-culture of about lOxlO 6 apoptotic cells is administered.
  • a dose derived from lOxlO 7 apoptotic cells is administered.
  • a dose derived from 10x10 apoptotic cells is administered.
  • a dose derived from lOxlO 9 apoptotic cells is administered.
  • a dose derived from lOxlO 10 apoptotic cells is administered.
  • a dose derived from lOxlO 11 apoptotic cells is administered.
  • apoptotic cell supernatant derived from the co-culture of about lOxlO 6 apoptotic cells is administered.
  • a dose derived from lOxlO 7 apoptotic cells is administered.
  • a dose derived from 10x10 apoptotic cells is administered.
  • a dose derived from 10x10 apoptotic cells is administered. In another embodiment, a dose derived from 10x10 5 apoptotic cells is administered. In another embodiment, a dose derived from lOxlO 4 apoptotic cells is administered. In another embodiment, a dose derived from 10x10 apoptotic cells is administered. In another embodiment, a dose derived from 10x10 2 apoptotic cells is administered.
  • a dose of apoptotic cell supernatant derived from 35xl0 6 apoptotic cells is administered.
  • a dose derived from 210xl0 6 apoptotic cells is administered.
  • a dose derived from 70x10 6 apoptotic cells is administered.
  • a dose derived from 140xl0 6 apoptotic cells is administered.
  • a dose derived from 35-210xl0 6 apoptotic cells is administered.
  • the apoptotic cell supernatant, or composition comprising said apoptotic cell supernatant may be administered by any method known in the art including, but not limited to, intravenous, subcutaneous, intranodal, intratumoral, intrathecal, intrapleural, intraperitoneal and directly to the thymus, as discussed in detail herein.
  • the apoptotic cell supernatants reduces production of cytokines associated with the cytokine storm such as IL-6.
  • Another cytokine, IL-2 is not involved in cytokine release syndrome although is secreted by DCs and macrophages in small quantities. It is, however, required for the survival and proliferation of CAR-T-cells and is mostly produced by these T-cells.
  • the apoptotic cell supernatants such as apoptotic cell-phagocyte supernatants, do not reduce IL-2 levels sufficiently to negatively affect the survival of CAR T-cells.
  • the apoptotic cell supernatants such as apoptotic cell-phagocyte supernatants, affect cytokine expression levels in macrophages and DCs, but do not affect cytokine expression levels in the T-cells themselves. It was therefore unexpected that apoptotic cell supernatants would be useful in enhancing CAR T-cell therapy or .
  • the apoptotic cell supernatants trigger death of T-cells, but not via changes in cytokine expression levels.
  • apoptotic cell supernatants such as apoptotic cell-phagocyte supernatants antagonize the priming of macrophages and dendritic cells to secrete cytokines that would otherwise amplify the cytokine storm.
  • apoptotic cell supernatants increase Tregs which suppress the inflammatory response and/or prevent excess release of cytokines.
  • administration of apoptotic cell supernatants inhibits one or more pro-inflammatory cytokines.
  • the pro-inflammatory cytokine comprises IL-lbeta, IL-6, TNF-alpha, or IFN-gamma, or any combination thereof.
  • administration of apoptotic cell supernatants promotes the secretion of one or more anti-inflammatory cytokines.
  • the anti- inflammatory cytokine comprises TGF-beta, IL10, or PGE2, or any combination thereof.
  • administration of apoptotic cell supernatants inhibits dendritic cell maturation following exposure to TLR ligands.
  • administration of apoptotic cell supernatants creates potentially tolerogenic dendritic cells, which in one embodiment, are capable of migration, and in one embodiment, the migration is due to CCR7.
  • administration of apoptotic cell supernatants elicits various signaling events which in one embodiment is TAM receptor signaling (Tyro3, Axl and Mer) which in one embodiment, inhibits inflammation in antigen-presenting cells.
  • Tyro-3, Axl, and Mer constitute the TAM family of receptor tyrosine kinases (RTKs) characterized by a conserved sequence within the kinase domain and adhesion molecule- like extracellular domains.
  • RTKs receptor tyrosine kinases
  • administration of apoptotic cell supernatants activates signaling through MerTK.
  • administration of apoptotic cell supernatants activates the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, which in one embodiment, negatively regulates NF- ⁇ .
  • PI3K phosphatidylinositol 3-kinase
  • administration of apoptotic cell supernatants negatively regulates the inflammasome which in one embodiment leads to inhibition of pro-inflammatory cytokine secretion, DC maturation, or a combination thereof.
  • administration of apoptotic cell supernatants upregulates expression of antiinflammatory genes such as Nr4a, Thbsl, or a combination thereof.
  • administration of apoptotic cell supernatants induces a high level of AMP which in one embodiment, is accumulated in a Pannexinl-dependent manner.
  • administration of apoptotic cell supernatants suppresses inflammation.
  • compositions for the treatment of a condition or disease as described herein are for maintaining or increasing the proliferation rate of a genetically modified immune cells.
  • methods for maintaining or increasing the proliferation rate of genetically modified immune cells further comprise reducing or inhibiting the incidence of cytokine release syndrome (CRS) or cytokine storm.
  • CRS cytokine release syndrome
  • disclosed herein are pharmaceutical compositions for increasing the efficacy of a genetically modified immune cell therapy.
  • compositions used in the methods for increasing the efficacy of an immune cell therapy further comprise reducing or inhibiting the incidence of CRS or a cytokine storm.
  • compositions for methods treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer of a tumor in a subject further comprise reducing or inhibiting the incidence of CRS or a cytokine storm.
  • a pharmaceutical composition comprises a genetically modified immune cell or a genetically modified receptor thereof.
  • a genetically modified immune cell comprises a T-cell.
  • a genetically modified immune cell comprises a chimeric antigen receptor CAR T-cell.
  • a genetically modified immune cell comprises a chimeric antigen receptor TCR T-cell.
  • a genetically modified immune cell comprises a cytotoxic T lymphocyte.
  • a genetically modified immune cell comprises a dendritic cell.
  • a genetically modified immune cell comprises a natural killer cell.
  • a genetically modified receptor comprises a genetically modified T-cell receptor.
  • a pharmaceutical composition for the treatment of a condition or a disease as described herein comprises an effective amount of a genetically modified immune cell or a genetically modified receptor thereof, as described herein in a pharmaceutically acceptable excipient.
  • a pharmaceutical composition for the treatment of a condition or a disease as described herein comprises an effective amount of a CAR T-cell as described herein in, and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition for the treatment of a condition or a disease as described herein comprises an effective amount of a TCR T-cell as described herein in, and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition for the treatment of a condition or a disease as described herein comprises an effective amount of a cytotoxic T-cell, as described herein, and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition for the treatment of a condition or a disease as described herein comprises an effective amount of a genetically modified dendritic cell, as described herein, and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition for the treatment of a condition or a disease as described herein comprises an effective amount of a genetically modified natural killer cell, as described herein, and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition for the treatment of a condition or a disease as described herein comprises an effective amount of a genetically modified T-cell receptor, as described herein, and a pharmaceutically acceptable excipient.
  • the condition or disease as described herein is a tumor or cancer.
  • a composition comprising the genetically modified immune cell or receptor thereof, for example a CAR T-cell, that binds to a protein or peptide of interest as described herein.
  • a composition comprising the genetically modified immune cell or receptor thereof, for example a TCR T-cell, that recognizes and binds a protein or peptide of interest as described herein.
  • the protein or peptide of interest comprises a tumor antigen or a fragment thereof.
  • a composition disclosed herein and used in methods disclosed herein comprises apoptotic cells or an apoptotic cell supernatant, and a pharmaceutically acceptable excipient.
  • a composition comprising an effective amount of a genetically modified immune cell or a genetically modified receptor thereof may be the same composition as comprises an apoptotic cell population or an apoptotic cell supernatant.
  • compositions comprising an effective amount of a CAR T-cell, or a TCR T-cell, or a cytotoxic T-cell, or a genetically modified dendritic cell, or a genetically modified natural killer cell may be the same composition as comprises an apoptotic cell population or an apoptotic cell supernatant.
  • a composition comprising an effective amount of genetically modified T-cell receptor may be the same composition as comprises an apoptotic cell population or an apoptotic cell supernatant.
  • a composition comprising an effective amount of a genetically modified immune cell selected from the group comprising a CAR T-cell, a TCR T-cell, a cytotoxic T-cell, a natural killer cell, or a dendritic cell, is not the same composition as comprises an apoptotic cell population or an apoptotic cell supernatant.
  • a composition comprises a chimeric antigen receptor-expressing T-cell (CAR T-cell) and either apoptotic cells or an apoptotic cell supernatant, and a pharmaceutically acceptable excipient.
  • a composition comprises a genetically modified T-cell receptor expressing T-cell (TCR T-cell) and either apoptotic cells or an apoptotic cell supernatant, and a pharmaceutically acceptable excipient.
  • TCR T-cell TCR T-cell
  • a composition comprising an effective amount of a genetically modified T-cell receptor is not the same composition as comprises an apoptotic cell population or an apoptotic cell supernatant.
  • apoptotic cells comprised in a composition comprise apoptotic cells in an early apoptotic state.
  • apoptotic cells comprised in a composition are pooled third party donor cells.
  • an apoptotic cell supernatant comprised in a composition disclosed herein is collected from early apoptotic cells.
  • an apoptotic cell supernatant comprised in a composition disclosed herein is collected pooled third party donor cells.
  • a composition comprising a genetically modified immune cells for example a CAR T-cell, further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells, for example a CAR T-cell, and apoptotic cells further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells, for example a CAR T-cell, and an apoptotic cell supernatant, further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a genetically modified immune cells for example a CAR T-cell
  • an apoptotic cell supernatant further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells, for example a TCR T-cell, and apoptotic cells further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells, for example a TCR T- cell, and an apoptotic cell supernatant, further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a genetically modified immune cells for example a TCR T- cell
  • an apoptotic cell supernatant further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells for example a dendritic cell, further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells, for example a dendritic, and apoptotic cells further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells, for example a dendritic, and an apoptotic cell supernatant, further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a genetically modified immune cells for example a dendritic
  • an apoptotic cell supernatant further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells for example a NK cell, further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells, for example a NK cell, and apoptotic cells further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a composition comprising a genetically modified immune cells, for example a NK cell, and an apoptotic cell supernatant, further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • a genetically modified immune cells for example a NK cell
  • an apoptotic cell supernatant further comprises an additional pharmaceutical composition for preventing, suppressing, or modulating cytokine release in a patient with cytokine release syndrome or experiencing a cytokine storm.
  • the additional pharmaceutical composition comprises a CTLA-4 blocking agent, which in one embodiment is Ipilimumab.
  • the additional pharmaceutical composition comprises a alpha- 1 anti- trypsin, as disclosed herein, or a fragment thereof, or an analogue thereof.
  • the additional pharmaceutical composition comprises a tellurium-based compound, a disclosed herein.
  • the additional pharmaceutical composition comprises an immune modulating agent, as disclosed herein.
  • the additional pharmaceutical composition comprises a CTLA-4 blocking agent, an alpha- 1 anti- trypsin or fragment thereof or analogue thereof, a tellurium-based compound, or an immune modulating compound, or any combination thereof.
  • the composition comprising the genetically modified immune cell for example a CAR T-cell and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, or an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent comprises a single composition.
  • the composition comprising the genetically modified immune cell for example CAR T-cells and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, or an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent, or any combination thereof, comprises multiple compositions, wherein each of the genetically modified immune cell, which in one embodiment is CAR T-cells, the CTLA-4 blocking agent, the alpha- 1 anti-trypsin or fragment thereof or analogue thereof, the apoptotic cells, the apoptotic cell supernatant, the tellurium-based compound, or the immune modulating agent, or any combination thereof, are comprised in a separate composition.
  • the composition comprising the genetically modified immune cell which in one embodiment is CAR T-cells and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent, or any combination thereof, comprises multiple compositions, wherein the genetically modified immune cells, which in one embodiment are CAR T-cells, the CTLA-4 blocking agent, or the alpha- 1 anti-trypsin or fragment thereof or analogue thereof, the tellurium-based compound, or the immune modulating agent, or any combination thereof, or any combination thereof are present in the genetically modified immune cell, for example a CAR T-cell, composition, and the apoptotic cells, or the apoptotic cell supernatant, are comprised in a separate composition
  • the composition comprising the genetically modified immune cell for example a TCR T-cell and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, or an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent comprises a single composition.
  • the composition comprising the genetically modified immune cell for example TCR T-cells and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, or an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent, or any combination thereof, comprises multiple compositions, wherein each of the genetically modified immune cell, which in one embodiment is TCR T-cells, the CTLA-4 blocking agent, the alpha- 1 anti-trypsin or fragment thereof or analogue thereof, the apoptotic cells, the apoptotic cell supernatant, the tellurium-based compound, or the immune modulating agent, or any combination thereof, are comprised in a separate composition.
  • the composition comprising the genetically modified immune cell which in one embodiment is TCR T- cells and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent, or any combination thereof, comprises multiple compositions, wherein the genetically modified immune cells, which in one embodiment are TCR T-cells, the CTLA-4 blocking agent, or the alpha- 1 anti-trypsin or fragment thereof or analogue thereof, the tellurium-based compound, or the immune modulating agent, or any combination thereof, or any combination thereof are present in the genetically modified immune cell, for example a TCR T-cell, composition, and the apoptotic cells, or the apoptotic cell supernatant, are comprised in a separate composition.
  • the composition comprising the genetically modified immune cell for example a dendritic cell and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, or an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent comprises a single composition.
  • the composition comprising the genetically modified immune cell for example dendritic cells and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, or an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent, or any combination thereof, comprises multiple compositions, wherein each of the genetically modified immune cell, which in one embodiment is dendritic cells, the CTLA-4 blocking agent, the alpha- 1 anti-trypsin or fragment thereof or analogue thereof, the apoptotic cells, the apoptotic cell supernatant, the tellurium-based compound, or the immune modulating agent, or any combination thereof, are comprised in a separate composition
  • the composition comprising the genetically modified immune cell which in one embodiment is dendritic cells and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-tryps
  • the composition comprising the genetically modified immune cell for example a NK cell and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analogue thereof, apoptotic cells, or an apoptotic cell supernatant, a tellurium-based compound, or an immune modulating agent comprises a single composition.
  • the composition comprising the genetically modified immune cell which in one embodiment is NK cells and the pharmaceutical composition comprising any one of a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or an
  • a "pharmaceutical composition” may encompass a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • physiologically acceptable carrier may encompass a carrier, excipient, or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered active ingredient.
  • excipient may encompass an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • compositions are administered at the same time. In an alternative embodiment, compositions are administered at different times. In another embodiment, compositions comprising apoptotic cells are administered prior to infusion or genetically modified immune cells or receptors thereof. In another embodiment, compositions comprising apoptotic cells are administered prior to CAR- T-cell infusion. In another embodiment, compositions comprising apoptotic cells are administered prior to cytotoxic T-cell infusion. In another embodiment, compositions comprising apoptotic cells are administered prior to natural killer cell infusion. In another embodiment, compositions comprising apoptotic cells are administered prior to dendritic infusion. In another embodiment, compositions comprising apoptotic cells are administered prior to infusion of a genetically modified T-cell receptor.
  • compositions comprising apoptotic cell supernatants are administered prior to infusion or genetically modified immune cells or receptors thereof.
  • compositions comprising apoptotic cell supernatants are administered prior to CAR- T-cell infusion.
  • compositions comprising apoptotic cell supernatants are administered prior to cytotoxic T-cell infusion.
  • compositions comprising apoptotic cell supernatants are administered prior to natural killer cell infusion.
  • compositions comprising apoptotic cell supernatants are administered prior to dendritic infusion.
  • compositions comprising apoptotic cell supernatants are administered prior to infusion of a genetically modified T-cell receptor.
  • compositions comprising apoptotic cell supernatants are administered prior to infusion of genetically modified immune cells or receptors thereof.
  • compositions comprising apoptotic cells are administered about 24 hours prior to genetically modified immune cell or receptor thereof infusion.
  • compositions comprising apoptotic cells are administered about 24 hours prior to CAR T-cell, or cytotoxic T-cells, or TCR T-cells, or natural killer cells, or dendritic cell or genetically modified T- cell receptor infusion.
  • compositions comprising apoptotic cell supernatants are administered about 24 hours prior to CAR T-cell or cytotoxic T-cells, or TCR T-cells, or natural killer cells, or dendritic cell or genetically modified T-cell receptor infusion.
  • compositions comprising apoptotic cells are administered about 2 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 18 hours 20 hours, 22 hours, 24 hours, 36 hours, 48 hours, 60 hours, or 72 hours prior to CAR- T-cell or cytotoxic T-cells, or TCR T-cells, or natural killer cells, or dendritic cell or genetically modified T-cell receptor infusion.
  • compositions comprising apoptotic cell supernatants are administered about 2 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 18 hours 20 hours, 22 hours, 24 hours, 36 hours, 48 hours, 60 hours, or 72 hours prior to CAR T-cell or cytotoxic T-cells, or TCR T- cells, or natural killer cells, or dendritic cell or genetically modified T-cell receptor infusion.
  • Each possibility represents a separate embodiment as disclosed herein.
  • compositions comprising apoptotic cells are administered after infusion of genetically modified immune cells or genetically modified receptors thereof.
  • composition comprising apoptotic cells are administered after CAR- T-cell or cytotoxic T-cells, or TCR T-cells, or natural killer cells, or dendritic cell or genetically modified T- cell receptor infusion.
  • compositions comprising apoptotic cell supernatants are administered after infusion of genetically modified immune cells or genetically modified receptors thereof.
  • compositions comprising apoptotic cell supernatants are administered after CAR T-cell or cytotoxic T-cells, or TCR T-cells, or natural killer cells, or dendritic cell or genetically modified T-cell receptor infusion.
  • compositions comprising apoptotic cells are administered about 24 hours after CAR-T-cell or cytotoxic T-cells, or TCR T-cells, or natural killer cells, or dendritic cell or genetically modified T-cell receptor infusion.
  • compositions comprising apoptotic cells are administered after infusion of genetically modified immune cells or genetically modified receptors thereof.
  • compositions comprising apoptotic cell supernatants are administered about 24 hours after CAR T-cell or cytotoxic T-cells, or TCR T-cells, or natural killer cells, or dendritic cell or genetically modified T-cell receptor infusion.
  • compositions comprising apoptotic cells are administered about 2 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 18 hours 20 hours, 22 hours, 24 hours, 36 hours, 48 hours, 60 hours, or 72 hours after CAR- T-cell or cytotoxic T-cells, or TCR T-cells, or natural killer cells, or dendritic cell or genetically modified T-cell receptor infusion.
  • compositions comprising apoptotic cell supernatants are administered about 2 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 18 hours 20 hours, 22 hours, 24 hours, 36 hours, 48 hours, 60 hours, or 72 hours after CAR T-cell or cytotoxic T-cells, or natural killer cells, or dendritic cell or genetically modified T-cell receptor infusion.
  • Each possibility represents a separate embodiment as disclosed herein.
  • compositions disclosed herein comprising genetically modified immunoresponsive cells or comprising the apoptotic cells or comprising the apoptotic cell supernatants, or any combination thereof, can be conveniently provided as sterile liquid preparations, e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions, which may be buffered to a selected pH, Liquid preparations are normally easier to prepare than gels, other viscous compositions, and solid compositions. Additionally, liquid compositions are somewhat more convenient to administer, especially by injection. Viscous compositions, on the other hand, can be formulated within the appropriate viscosity range to provide longer contact periods with specific tissues.
  • Liquid or viscous compositions can comprise carriers, which can be a solvent or dispersing medium containing, for example, water, saline, phosphate buffered saline, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like) and suitable mixtures thereof.
  • carriers can be a solvent or dispersing medium containing, for example, water, saline, phosphate buffered saline, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like) and suitable mixtures thereof.
  • Sterile injectable solutions can be prepared by incorporating the genetically modified immunoresponsive cells or apoptotic cell supernatants utilized in practicing the methods disclosed herein, in the required amount of the appropriate solvent with various amounts of the other ingredients, as desired.
  • Such compositions may be in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose, dextrose, or the like.
  • the compositions can also be lyophilized.
  • compositions can contain auxiliary substances such as wetting, dispersing, or emulsifying agents (e.g., methylcellulose), pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • auxiliary substances such as wetting, dispersing, or emulsifying agents (e.g., methylcellulose), pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • Standard texts such as "REMINGTON'S PHARMACEUTICAL SCIENCE", 17th edition, 1985, incorporated herein by reference, may be consulted to prepare suitable preparations, without undue experimentation.
  • compositions which enhance the stability and sterility of the compositions, including antimicrobial preservatives, antioxidants, chelating agents, and buffers, can be added.
  • Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. According to the disclosure herein, however, any vehicle, diluent, or additive used would have to be compatible with the genetically modified immunoresponsive cells or their progenitors.
  • compositions can be isotonic, i.e., they can have the same osmotic pressure as blood and lacrimal fluid.
  • the desired isotonicity of the compositions as disclosed herein may be accomplished using sodium chloride, or other pharmaceutically acceptable agents such as dextrose, boric acid, sodium tartrate, propylene glycol or other inorganic or organic solutes.
  • Sodium chloride may be preferred particularly for buffers containing sodium ions.
  • Viscosity of the compositions can be maintained at the selected level using a pharmaceutically acceptable thickening agent.
  • Methylcellulose may be preferred because it is readily and economically available and is easy to work with.
  • suitable thickening agents include, for example, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, carbomer, and the like.
  • concentration of the thickener will depend upon the agent selected. The important point is to use an amount that will achieve the selected viscosity.
  • suitable carriers and other additives will depend on the exact route of administration and the nature of the particular dosage form, e.g., liquid dosage form (e.g., whether the composition is to be formulated into a solution, a suspension, gel or another liquid form, such as a time release form or liquid-filled form).
  • compositions should be selected to be chemically inert and will not affect the viability or efficacy of the genetically modified immunoresponsive cells as described in the methods disclosed herein. This will present no problem to those skilled in chemical and pharmaceutical principles, or problems can be readily avoided by reference to standard texts or by simple experiments (not involving undue experimentation), from this disclosure and the documents cited herein.
  • One consideration concerning the therapeutic use of genetically modified immunoresponsive cells disclosed herein is the quantity of cells necessary to achieve an optimal effect.
  • the quantity of cells to be administered will vary for the subject being treated. In a one embodiment, between 10 4 to 10 10 , between 10 5 to 10 9 , or between 10 6 and 10 8 genetically modified immunoresponsive cells disclosed herein are administered to a human subject. More effective cells
  • 3x10 , 4 x 10 , and 5 x 10 genetically modified immunoresponsive cells disclosed herein are administered to a human subject.
  • the precise determination of what would be considered an effective dose may be based on factors individual to each subject, including their size, age, sex, weight, and condition of the particular subject. Dosages can be readily ascertained by those skilled in the art from this disclosure and the knowledge in the art.
  • any additives in addition to the active cell(s) and/or agent(s) are present in an amount of 0.001 to 50% (weight) solution in phosphate buffered saline, and the active ingredient is present in the order of micrograms to milligrams, such as about 0.0001 to about 5 wt %. In another embodiment about 0.0001 to about 1 wt %. In still another embodiment, about 0.0001 to about 0.05 wt% or about 0.001 to about 20 wt %. In a further embodiment, about 0.01 to about 10 wt %.
  • 0.05 to about 5 wt % In another embodiment, about 0.05 to about 5 wt %.
  • toxicity such as by determining the lethal dose (LD) and LD50 in a suitable animal model e.g., rodent such as mouse; and, the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response.
  • LD lethal dose
  • LD50 LD50
  • a suitable animal model e.g., rodent such as mouse
  • a nucleic acid sequence encoding a chimeric antigen receptor (CAR) as described herein for uses in the compositions and methods as disclosed herein.
  • a vector comprising the nucleic acid sequence encoding a chimeric antigen receptor (CAR) as described herein.
  • a vector comprising the nucleic acid sequence encoding a genetically modified T-cell receptor (TCR) as described herein.
  • TCR genetically modified T-cell receptor
  • Genetic modification of immunoresponsive cells can be accomplished by transducing a substantially homogeneous cell composition with a recombinant DNA construct.
  • a retroviral vector (either gamma- retroviral or lentiviral) is employed for the introduction of the DNA construct into the cell.
  • a polynucleotide encoding a receptor that binds an antigen e.g., a tumor antigen, or a valiant, or a fragment thereof
  • an antigen e.g., a tumor antigen, or a valiant, or a fragment thereof
  • Non- viral vectors may be used as well.
  • Non- viral approaches can also be employed for the expression of a protein in cell.
  • a nucleic acid molecule can be introduced into a cell by administering the nucleic acid in the presence of lipofection (Feigner et al., Proc. Natl. Acad. Sci. U.S.A. 84:7413, 1987; Ono et al., Neuroscience Letters 17:259, 1990; Brigham et al, Am. J. Med. Sci.
  • Transplantation of normal genes into the affected tissues of a subject can also be accomplished by transferring a normal nucleic acid into a cultivatable cell type ex vivo (e.g., an autologous or heterologous primary cell or progeny thereof), after which the cell (or its descendants) are injected into a targeted tissue or are injected systemically.
  • Recombinant receptors can also be derived or obtained using transposases or targeted nucleases (e.g. Zinc finger nucleases, meganucleases, or TALE nucleases).
  • Transient expression may be obtained by RNA electroporation.
  • cDNA expression for use in polynucleotide therapy methods can be directed from any suitable promoter (e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metallothionein promoters), and regulated by any appropriate mammalian regulatory element or intron (e.g. the elongation factor la enhancer/promoter/intron structure).
  • CMV human cytomegalovirus
  • SV40 simian virus 40
  • metallothionein promoters regulated by any appropriate mammalian regulatory element or intron (e.g. the elongation factor la enhancer/promoter/intron structure).
  • enhancers known to preferentially direct gene expression in specific cell types can be used to direct the expression of a nucleic acid.
  • the enhancers used can include, without limitation, those that are characterized as tissue- or cell- specific enhancers.
  • regulation can be mediated by the cognate regulatory sequences or, if desired, by regulatory sequences derived from a heterologous source, including any of the promoters or regulatory elements described above.
  • a cell comprising the vector comprising the nucleic acid sequence encoding a chimeric antigen receptor (CAR) as disclosed herein
  • a cell comprising the vector comprising the nucleic acid sequence encoding a genetically modified T-cell receptor (TCR) as disclosed herein.
  • CAR chimeric antigen receptor
  • TCR genetically modified T-cell receptor
  • kits for treatment of a neoplasia, pathogen infection, an autoimmune disorder, or an allogeneic transplant comprising a CAR T-cells and apoptotic cells as disclosed herein, either separately or pre-mixed.
  • kits for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject comprising a CAR T-cells and apoptotic cells as disclosed herein, either separately or pre-mixed.
  • a kit for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject the kit comprising a CAR T-cells and an apoptotic cell supernatant as disclosed herein, either separately or pre-mixed.
  • kits for treatment of a neoplasia, pathogen infection, an autoimmune disorder, or an allogeneic transplant comprising a TCR T-cells and apoptotic cells as disclosed herein, either separately or pre-mixed.
  • kits for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject comprising a TCR T-cells and apoptotic cells as disclosed herein, either separately or pre-mixed.
  • a kit for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor in a subject the kit comprising a TCR T-cells and an apoptotic cell supernatant as disclosed herein, either separately or pre-mixed.
  • kits for the treatment or prevention of a neoplasia, pathogen infection, immune disorder or allogeneic transplant, or for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor includes a therapeutic or prophylactic composition containing an effective amount of an immunoresponsive cells and apoptotic cells as disclosed herein in unit dosage form.
  • the kit includes a therapeutic or prophylactic composition containing an effective amount of an immunoresponsive cells and an apoptotic cell supernatant as disclosed herein in unit dosage form.
  • the cells further comprise a co-stimulatory ligand.
  • kits further comprise an additional agent selected from the group comprising a CTLA- 4 blocking agent, an alpha- 1 anti-trypsin or fragment thereof or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof.
  • the kit comprises a sterile container which contains a therapeutic or prophylactic vaccine; such containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art.
  • Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding medicaments.
  • the immunoresponsive cells and apoptotic cells or apoptotic cell supernatant are provided together with instructions for administering the cells to a subject having or at risk of developing a neoplasia, pathogen infection, immune disorder or allogeneic transplant or tumors or cancer.
  • the instructions will generally include information about the use of the composition for the treatment or prevention of neoplasia, pathogen infection, immune disorder, allogeneic transplant, tumor or cancer.
  • the instructions include at least one of the following: description of the therapeutic agent; dosage schedule and administration for treatment or prevention of a neoplasia, pathogen infection, immune disorder or allogeneic transplant, cancers, tumors, or symptoms thereof; precautions; warnings; indications; counter-indications; over dosage information; adverse reactions; animal pharmacology; clinical studies; and/or references.
  • the instructions may be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container.
  • antigen recognizing receptor may encompass a receptor that is capable of activating an immune cell (e.g., a T-cell) in response to antigen binding.
  • exemplary antigen recognizing receptors may be native or endogenous T-cell receptors or chimeric antigen receptors in which a tumor antigen-binding domain is fused to an intracellular signaling domain capable of activating an immune cell (e.g., a T-cell).
  • antibody means not only intact antibody molecules, but also fragments of antibody molecules that retain immunogen-binding ability. Such fragments are also well known in the art and are regularly employed both in vitro and in vivo. Accordingly, the skilled artisan would appreciate that the term “antibody” means not only intact immunoglobulin molecules but also the well-known active fragments F(ab 1 )2, and Fab. F(ab')2 5 and Fab fragments that lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding of an intact antibody (Wahl et al., J. Nucl. Med. 24:316-325 (1983).
  • the antibodies disclosed herein comprise whole native antibodies, bispecific antibodies; chimeric antibodies; Fab, Fab', single chain V region fragments (scFv), fusion polypeptides, and unconventional antibodies.
  • single -chain variable fragment encompasses a fusion protein of the variable regions of the heavy (VH) and light chains (VL) of an immunoglobulin covalently linked to form a VH::VL heterodimer.
  • the heavy (VH) and light chains (VL) are either joined directly or joined by a peptide-encoding linker (e.g., 30, 15, 20, 25 amino acids), which connects the N-terminus of the VH with the C-terminus of the VL, or the C-terminus of the VH with the N-terminus of the VL,
  • the linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility Despite removal of the constant regions and the introduction of a linker, scFv proteins retain the specificity of the original immunoglobulin.
  • Single chain Fv polypeptide antibodies can be expressed from a nucleic acid including VH- and VL-encoding sequences as described by Huston, et al.
  • Antagonistic scFvs having inhibitory activity have been described (see, e.g., Zhao et al., Hyrbidoma (Larchmt) 2008 27(6):455-51 ; Peter et al., J Cachexia Sarcope ia Muscle 2012 August 12; Shieh et al, J Imunol2009 183(4):2277-85; Giomarelli et al, Thromb Haemost 2007 97(6):955-63; Fife eta., J Clin Invst 2006 1 16(8):2252-61 ; Brocks et al, Immunotechnology 1997 3(3):173-84; Moosmayer et al, Ther Immunol 1995 2(10:31-40).
  • affinity is meant a measure of binding strength. Without being bound to theory, affinity depends on the closeness of stereochemical fit between antibody combining sites and antigen determinants, on the size of the area of contact between them, and on the distribution of charged and hydrophobic groups. Affinity also includes the term “avidity,” which refers to the strength of the antigen-antibody bond after formation of reversible complexes. Methods for calculating the affinity of an antibody for an antigen are known in the art, including use of binding experiments to calculate affinity. Antibody activity in functional assays (e.g., flow cytometry assay) is also reflective of antibody affinity. Antibodies and affinities can be phenotypically characterized and compared using functional assays (e.g., flow cytometry assay).
  • chimeric antigen receptor may encompass an antigen-binding domain that is fused to an intracellular signaling domain capable of activating or stimulating an immune cell.
  • the CAR's extracellular binding domain is composed of a single chain variable fragment (scFv) derived from fusing the variable heavy and light regions of a murine or humanized monoclonal antibody.
  • scFvs may be used that are derived from Fab's (instead of from an antibody, e.g., obtained from Fab libraries), in various embodiments, this scFv is fused to a transmembrane domain and then to an intracellular signaling domain.
  • the CAR is selected to have high affinity or avidity for the antigen.
  • polypeptides and Analogs are also included in the methods disclosed herein.
  • the methods disclosed herein comprise optimizing an amino acid sequence or nucleic acid sequence by producing an alteration in the sequence. Such alterations may include certain mutations, deletions, insertions, or post-translational modifications.
  • the disclosure provided herein further includes analogs of any naturally-occurring polypeptide disclosed herein.
  • Analogs can differ from a naturally-occurring polypeptide disclosed herein by amino acid sequence differences, by post-translational modifications, or by both. Analogs disclosed herein will generally exhibit at least 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%>, 99% or more identity with all or part of a naturally-occurring amino, acid sequence disclosed herein.
  • the length of sequence comparison is at least 5, 10, 15 or 20 amino acid residues. In another embodiment, at least 25, 50, or 75 amino acid residues. In still another embodiment, more than 100 amino acid residues.
  • a BLAST program may be used, with a probability score between e"3 and e"100 indicating a closely related sequence.
  • Modifications include in vivo and in vitro chemical derivatization of polypeptides, e.g., acetylation, carboxylation, phosphorylation, or glycosylation; such modifications may occur during polypeptide synthesis or processing or following treatment with isolated modifying enzymes. Analogs can also differ from the naturally-occurring polypeptides disclosed herein by alterations in primary sequence.
  • Non-protein analogs have a chemical structure designed to mimic the functional activity of a protein disclosed herein. Such analogs are administered according to methods disclosed herein. Such analogs may exceed the physiological activity of the original polypeptide. Methods of analog design are well known in the art, and synthesis of analogs can be carried out according to such methods by modifying the chemical structures such that the resultant analogs increase the antineoplastic activity of the original polypeptide when expressed in an immunoresponsive cell. These chemical modifications include, but are not limited to, substituting alternative R groups and varying the degree of saturation at specific carbon atoms of a reference polypeptide. In another embodiment, the protein analogs are relatively resistant to in vivo degradation, resulting in a more prolonged therapeutic effect upon administration. Assays for measuring functional activity include, but are not limited to, those described in the Examples below.
  • immunosuppressive activity describes induction of signal transduction or changes in protein expression in a cell (e.g., an activated immunoresponsive cell) resulting in a decrease in an immune response.
  • Polypeptides known to suppress or decrease an immune response via their binding include CD47, PD-1 , CTLA-4, and their corresponding ligands, including SIRPa, PD-L1, PD-L2, B7-1, and B7-2.
  • Such polypeptides are present in the tumor microenvironment and inhibit immune responses to neoplastic cells.
  • inhibiting, blocking, or antagonizing the interaction of immunosuppressive polypeptides and/or their ligands enhances the immune response of the immunoresponsive cell.
  • immunoresponsive activity describes induction of signal transduction or changes in protein expression in a cell (e.g., an activated immunoresponsive cell) resulting in an increased immune response.
  • Immunostimulatory activity may include pro-inflammatory activity.
  • Polypeptides known to stimulate or increase an immune response via their binding include CD28, OX-40, 4- IBB, and their corresponding ligands, including B7-1 , B7-2, OX-40L, and 4-1BBL.
  • Such polypeptides are present in the tumor microenvironment and activate immune responses to neoplastic cells.
  • promoting, stimulating, or agonizing pro -inflammatory polypeptides and/or their ligands enhances the immune response of the immunoresponsive cell.
  • Nucleic acid molecules useful in the methods disclosed herein include any nucleic acid molecule that encodes a polypeptide disclosed herein or a fragment thereof. Such nucleic acid molecules need not be 100% identical with an endogenous nucleic acid sequence, but will typically exhibit substantial identity. Polynucleotides having "substantial identity" to an endogenous sequence are typically capable of hybridizing with at least one strand of a double-stranded nucleic acid molecule. By “hybridize” is meant pair to form a double- stranded molecule between complementary polynucleotide sequences (e.g., a gene described herein), or portions thereof, under various conditions of stringency. (See, e.g., Wahl, G. M. and S. L. Berger (1987) Methods Enzymol. 152:399; immel, A. R. (1987) Methods Enzymol. 152:507).
  • substantially identical may encompass a polypeptide or nucleic acid molecule exhibiting at least 50% identity to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein). In one embodiment, such a sequence is at least 60%, 80% or 85%, 90%, 95% or even 99% identical at the amino acid level or nucleic acid to the sequence used for comparison.
  • Sequence identity is typically measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. In an exemplary approach to determining the degree of identity, a BLAST program may be used, with a probability score between e-3 and e- 100 indicating a closely related sequence.
  • sequence analysis software for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center
  • analog may encompass a structurally related polypeptide or nucleic acid molecule having the function of a reference polypeptide or nucleic acid molecule.
  • ligand may encompass a molecule that binds to a receptor.
  • the ligand binds a receptor on another cell, allowing for cell-to-cell recognition and/or interaction.
  • disease ay encompass any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ. Examples of diseases include neoplasia or pathogen infection of cell.
  • an "effective amount” may encompass an amount sufficient to have a therapeutic effect.
  • an “effective amount” is an amount sufficient to arrest, ameliorate, or inhibit the continued proliferation, growth, or metastasis (e.g., invasion, or migration) of a neoplasia.
  • Neoplasia may encompass a disease characterized by the pathological proliferation of a cell or tissue and its subsequent migration to or invasion of other tissues or organs. Neoplasia growth is typically uncontrolled and progressive, and occurs under conditions that would not elicit, or would cause cessation of, multiplication of normal cells.
  • Neoplasias can affect a variety of cell types, tissues, or organs, including but not limited to an organ selected from the group consisting of bladder, bone, brain, breast, cartilage, glia, esophagus, fallopian tube, gallbladder, heart, intestines, kidney, liver, lung, lymph node, nervous tissue, ovaries, pancreas, prostate, skeletal muscle, skin, spinal cord, spleen, stomach, testes, thymus, thyroid, trachea, urogenital tract, ureter, urethra, uterus, and vagina, or a tissue or cell type thereof.
  • Neoplasias include cancers, such as sarcomas, carcinomas, or plasmacytomas (malignant tumor of the plasma cells).
  • pathogen may encompass a virus, bacteria, fungi, parasite or protozoa capable of causing disease.
  • tumor antigen or “tumor associated antigen” may encompass an antigen (e.g., a polypeptide) that is uniquely or differentially expressed on a tumor cell compared to a normal or non- IS neoplastic cell.
  • a tumor antigen includes any polypeptide expressed by a tumor that is capable of activating or inducing an immune response via an antigen recognizing receptor (e.g., CD 19, MUCI) or capable of suppressing an immune response via receptor-ligand binding (e.g., CD47, PD-Ll/L2, B7.1/2).
  • virus antigen may encompass a polypeptide expressed by a virus that is capable of inducing an immune response.
  • compositions and methods as disclosed herein are envisioned to either comprise the active ingredient or specified step, consist of the active ingredient or specified step, or consist essentially of the active ingredient or specified step.
  • treatment may encompass clinical intervention in an attempt to alter the disease course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology.
  • Therapeutic effects of treatment include, without limitation, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastases, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • a treatment can prevent deterioration due to a disorder in an affected or diagnosed subject or a subject suspected of having the disorder, but also a treatment may prevent the onset of the disorder or a symptom of the disorder in a subject at risk for the disorder or suspected of having the disorder.
  • subject may encompass a vertebrate, in one embodiment, to a mammal, and in one embodiment, to a human. Subject may also refer, in one embodiment, to domesticated such as cows, sheep, horses, cats, dogs and laboratory animals such as mice, rats, gerbils, hamsters, etc.
  • TCR T-cells in which the TCR is directed to a peptide of interest.
  • the TCR binds to a peptide of interest.
  • the TCR recognizes a peptide of interest.
  • the TCR is a ligand of the peptide of interest.
  • the peptide of interest is a ligand of the TCR.
  • the immune cell as disclosed herein is not a T-cell. In another embodiment, the immune cell as disclosed herein is not an NK cell. In another embodiment, the immune cell as disclosed herein is not a CTL. In another embodiment, the immune cell as disclosed herein is not a regulatory T-cell. In another embodiment, the immune cell is not a human embryonic stem cell. In another embodiment, the immune cell is not a pluripotent stem cell from which lymphoid cells may be differentiated.
  • One approach to immunotherapy involves engineering a patient's own immune cells to create genetically modified immune cells that will recognize and attack their tumor.
  • Immune cells are collected and genetically modified, as described herein, for example to produce chimeric antigen receptors (CAR) on their cell surface that will allow the immune cell, for example a T-cell, to recognize a specific protein antigen on a tumor or cancer cell.
  • An expanded population of genetically modified immune cells for example CAR T-cells, is then administered to the patient.
  • the administered cells multiply in the patient' s body and recognize and kill cancer and tumor cells that harbor the antigen on their surface.
  • the administered cells multiply in a patient's body and recognize and kill tumor-associated antigens, which leads to the death of cancer and tumor cells.
  • disclosed herein are methods for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor comprising the step of administering a composition as disclosed herein.
  • a method for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor comprising the step of administering genetically modified immune cells and a composition comprising an additional agent, wherein said additional agent comprises apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, wherein said method treats, prevents, inhibits, reduces the incidence of, ameliorates or alleviates a cancer or a tumor in said subject compared with a subject administered said genetically modified immune cells and not administered the additional agent.
  • said genetically modified immune cells comprise genetically modified T-cell, cytotoxic T-cells, Treg cells, effector T-cells, helper T-cells, NK cells, or dendritic cells.
  • chimeric antigen receptor-expressing T-cells CAR T-cells
  • a composition comprising an additional agent, wherein said additional agent comprises apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof
  • said method treats, prevents, inhibits, reduces the incidence of, ameliorates or alleviates a cancer or a tumor in said subject compared with a subject administered said genetically modified immune cells and not administered the additional agent.
  • TCR T-cells genetically modified T-cell receptor cells
  • a composition comprising an additional agent, wherein said additional agent comprises apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof
  • said method treats, prevents, inhibits, reduces the incidence of, ameliorates or alleviates a cancer or a tumor in said subject compared with a subject administered said genetically modified immune cells and not administered the additional agent.
  • administration of apoptotic cells or an apoptotic supernatant or compositions thereof does not reduce the efficacy for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor, of said administering chimeric antigen receptor-expressing T-cells.
  • an additional agent comprising apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof does not reduce the efficacy for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor, of said administering chimeric antigen receptor-expressing T-cells.
  • administration of apoptotic cells or an apoptotic supernatant or compositions thereof increases the efficacy for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor, of said administering chimeric antigen receptor-expressing T-cells.
  • an additional agent comprising apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof increases the efficacy for treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or a tumor, of said administering chimeric antigen receptor-expressing T-cells.
  • methods increasing the efficacy of a genetically modified immune cell cancer therapy comprise administering said genetically modified immune cells and an additional agent comprising apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof, wherein the efficacy is increased compared with a subject not administered said additional agent.
  • said genetically modified immune cells are T-cells.
  • a T-cell is a naive T- cell.
  • a T-cell is a naive CD4 + T-cell.
  • a T-cell is a naive T-cell. In another embodiment, a T-cell is a naive CD8 + T-cell. In another embodiment, the genetically modified immune cell is a natural killer (NK) cell. In another embodiment, the genetically modified immune cell is a dendritic cell. In still another embodiment, the genetically modified T-cell is a cytotoxic T lymphocyte (CTL cell). In another embodiment, the genetically modified T-cell is a regulatory T-cell (Treg). In another embodiment, the genetically modified T- cell is a chimeric antigen receptor (CAR) T-cell. In another embodiment, the genetically modified T-cell is a genetically modified T-cell receptor cell (TCR T-cell).
  • NK natural killer
  • the genetically modified immune cell is a dendritic cell.
  • the genetically modified T-cell is a cytotoxic T lymphocyte (CTL cell).
  • CTL cell cytotoxic T lymphocyte
  • the genetically modified T-cell is a regulatory
  • methods increasing the efficacy of a CAR T-cell cancer therapy comprise administering said genetically modified immune cells and an additional agent comprising apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof, wherein the efficacy is increased compared with a subject not administered said additional agent.
  • methods herein reduce the level of production of at least one proinflammatory cytokine compared with the level of said pro-inflammatory cytokine in a subject receiving an immune cancer therapy and not administered an additional agent.
  • methods herein inhibit or reduce the incidence of cytokine release syndrome or cytokine storm in a subject undergoing a genetically modified immune cell cancer therapy and not administered an additional agent.
  • methods disclosed herein reduce IL-6.
  • methods herein increase the production of at least one cytokine compared with the level of said cytokine in a subject receiving an immune cancer therapy and not administered an additional agent.
  • the additional agent is apoptotic cells, In other embodiment, the additional agent is an apoptotic cell supernatant. In another embodiment, methods disclosed herein increase IL-2.
  • production may encompass expression of the cytokine as well as secretion of the cytokine from a cell.
  • increased production of a cytokine results in increased secretion of the cytokine from the cell.
  • decreased production of a cytokine results in decreased secretion of the cytokine from the cell.
  • methods disclosed herein decrease secretion of at least one cytokine.
  • methods disclosed herein decrease secretion of IL-6.
  • methods disclosed herein increase secretion of at least one cytokine.
  • methods disclosed herein increase secretion of IL-2.
  • a cell secreting at least one cytokine is a tumor cell. In another embodiment, a cell secreting at least one cytokine is a T-cell. In another embodiment, a cell secreting at least one cytokine is an immune cell. In another embodiment, a cell secreting at least one cytokine is a macrophage. In another embodiment, a cell secreting at least one cytokine is a B cell lymphocyte. In another embodiment, a cell secreting at least one cytokine is a mast cell. In another embodiment, a cell secreting at least one cytokine is an endothelial cell. In another embodiment, a cell secreting at least one cytokine is a fibroblast. In another embodiment, a cell secreting at least one cytokine is a stromal cell. A skilled artisan would recognize that the level of cytokines may be increased or decreased in cytokine secreting cells depending on the environment surrounding the cell.
  • an additional agent used in the methods disclosed herein increases secretion of at least one cytokine. In yet another embodiment, an additional agent used in the methods disclosed herein maintains secretion of at least one cytokine. In still another embodiment, an additional agent used in the methods disclosed herein does not decrease secretion of at least one cytokine. In another embodiment, an additional agent used in the methods disclosed herein increases secretion of IL-2. In another embodiment, an additional agent used in the methods disclosed herein increases secretion of IL-2R. In another embodiment, an additional agent used in the methods disclosed herein maintains secretion levels of IL-2. In another embodiment, an additional agent used in the methods disclosed herein maintains secretion levels of IL-2R.
  • an additional agent used in the methods disclosed herein does not decrease secretion levels of IL-2R. In another embodiment, an additional agent used in the methods disclosed herein maintains or increases secretion levels of IL-2. In another embodiment, an additional agent used in the methods disclosed herein maintains or increases secretion levels of IL- 2R. In another embodiment, an additional agent used in the methods disclosed herein does not decrease secretion levels of IL-2R.
  • an additional agent used in the methods disclosed herein decreases secretion of IL-6.
  • an additional agent used in the methods disclosed herein maintains, increases, or does not decrease secretion levels of IL-2 while decreasing secretion of IL-6.
  • an additional agent used in the methods disclosed herein maintains, increases, or does not decrease secretion levels of IL-2R while decreasing secretion of IL-6.
  • methods of increasing the efficacy of a CAR T-cell cancer therapy disclosed herein comprises decreasing the level of IL-6 in said subject, said method comprising administering CAR T-cells and an additional agent comprising apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti- trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof, wherein the efficacy is increased compared with a subject not administered said additional agent.
  • methods of increasing the efficacy of a CAR T-cell cancer therapy disclosed herein comprises increasing the level of IL-2 in said subject, said method comprising administering CAR T-cells and an additional agent comprising apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof, wherein the efficacy is increased compared with a subject not administered said additional agent.
  • methods of increasing the efficacy of a CAR T-cell cancer therapy disclosed herein comprises increasing proliferation of said CAR T-cells, said method comprising administering CAR T-cells and an additional agent comprising apoptotic cells, a supernatant from apoptotic cells, a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof, wherein the efficacy and proliferation of said CAR T- cells is increased compared with a subject not administered said additional agent.
  • methods of increasing the efficacy of CAR T-cell cancer therapy, decrease or inhibit cytokine production in the subject comprising the step of administering a composition comprising CAR T-cells and a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof.
  • methods of treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or tumor also decrease or inhibit cytokine production in the subject, said methods comprising the step of administering a composition comprising CAR T-cells and a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium-based compound, or an immune modulating agent, or any combination thereof, or compositions thereof.
  • cytokine release syndrome or cytokine storm in a subject undergoing CAR T-cell cancer therapy.
  • methods of treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a cancer or tumor decrease or inhibit cytokine production in a subject, said methods comprising the step of administering a composition comprising CAR T-cells and a CTLA-4 blocking agent, an alpha- 1 anti-trypsin or fragment or analog thereof, a tellurium- based compound, or an immune modulating agent, or any combination thereof, or compositions thereof.
  • disclosed herein are methods of preventing cytokine release syndrome or cytokine storm in a subject undergoing CAR T-cell cancer therapy. In another embodiment, disclosed herein are methods of alleviating cytokine release syndrome or cytokine storm in a subject undergoing CAR T-cell cancer therapy. In another embodiment, disclosed herein are methods of ameliorating cytokine release syndrome or cytokine storm in a subject undergoing CAR T-cell cancer therapy. In another embodiment, administration of apoptotic cells or an apoptotic supernatant or compositions thereof does not reduce the efficacy of the CAR T-cell therapy.
  • cytokine release syndrome CRS
  • CAR T-cell chimeric antigen receptor-expressing T-cell
  • the method comprises the step of administering a composition comprising apoptotic cells or an apoptotic cell supernatant or compositions thereof to said subject.
  • inhibiting or reducing the incidence of a cytokine release syndrome (CRS) or a cytokine storm is determined by measuring cytokine levels in a subject undergoing chimeric antigen receptor-expressing T-cell cancer therapy and being administered apoptotic cells or an apoptotic supernatant.
  • measured levels of cytokines are compared with cytokine levels in a subject not undergoing CAR T-cell cancer therapy.
  • measured cytokine levels are compared with cytokine levels in a subject not administer apoptotic cells or an apoptotic supernatant.
  • measured cytokine levels are compared with a control subject.
  • the level of pro-inflammatory cytokines are reduced in the subject compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant or compositions thereof.
  • methods disclosed herein reduce or inhibit the level of production of at least one pro-inflammatory cytokines compared with a subject undergoing CAR T-cell cancer therapy and not administered said apoptotic cells or said apoptotic cell supernatant or compositions thereof.
  • a method disclosed herein may further comprise administration of additional agents.
  • a method disclosed herein may comprise administration of additional agents and not apoptotic cells or an apoptotic cell supernatant.
  • additional agents may be those compounds or compositions that maintain, enhance, or improve, or any combination thereof, CAR T-cell cancer therapy.
  • additional agents that maintain, enhance, or improve CAR T-cell cancer therapy include CTLA-4 blocking agents, an alpha- lanti-trypsin or functional fragment thereof, or an analogue thereof, a tellurium-based compound, or an immune-modulating drug, or any combination thereof.
  • an additional agent includes apoptotic cells or an apoptotic supernatant.
  • administration of an additional agent, a described herein is prior to, concurrent with, of following said CAR T-cell cancer therapy.
  • an IL-6 receptor antagonist which in one embodiment is tocilizumab is used with the compositions and methods as disclosed herein.
  • adoptively transferred T-cells engraft and expand more efficiently in a lymphopenic host.
  • the subject is subjected to lymphodepletion prior to transfer of CAR T-cells or other modified immune cells.
  • the subject receiving the CAR T-cells is given T-cell-supportive cytokines.
  • the T-cells are effector T-cells.
  • the T-cells are naive T-cells.
  • the T-cells are central memory (T C M) T-cells.
  • the T-cells are Thl7 cells.
  • the T-cells are T stem memory cells.
  • the T-cells are regulatory T-cells.
  • the T-cells are cytotoxic T-cells.
  • the T-cells have high replicative capacity.
  • T-cell expansion occurs in the patient.
  • small numbers of cells may be transferred to a patient.
  • T-cell expansion occurs in vitro.
  • large numbers of cells may be transferred to a patient, cells and/or supernatants may be transferred to a patient on multiple occasions, or a combination thereof.
  • an advantage of CAR T-cells is that because they are specific for cell- surface molecules, they overcome the constraints of MHC-restricted TCR recognition and avoid tumor escape through impairments in antigen presentation or human leukocyte antigen expression.
  • reducing the tumor burden comprises reducing the number of tumor cells in the subject.
  • reducing the tumor burden comprises reducing tumor size in the subject.
  • reducing the tumor burden comprises eradicating the tumor in the subject.
  • a method of inducing tumor cell death in a subject comprising the step of administering to said subject any of the compositions as described herein.
  • a method as disclosed herein for inducing tumor cell death in a subject comprises administering immune cells, such as NK cells or T-cells comprising engineered chimeric antigen receptors with at least an additional agent to decrease toxic cytokine release or "cytokine release syndrome” (CRS) or "severe cytokine release syndrome” (sCRS) or "cytokine storm" in the subject.
  • immune cells such as NK cells or T-cells comprising engineered chimeric antigen receptors with at least an additional agent to decrease toxic cytokine release or "cytokine release syndrome” (CRS) or "severe cytokine release syndrome” (sCRS) or "cytokine storm" in the subject.
  • CRS cytokine release syndrome
  • sCRS severe cytokine release syndrome
  • a method of increasing or lengthening the survival of a subject having neoplasia comprising the step of administering to said subject any of the compositions as described herein.
  • a method of increasing or lengthening the survival of a subject comprises administering immune cells, such as NK cells or T- cells comprising engineered chimeric antigen receptors with at least an additional agent to decrease toxic cytokine release or "cytokine release syndrome” (CRS) or "severe cytokine release syndrome” (sCRS) or "cytokine storm" in the subject.
  • immune cells such as NK cells or T- cells comprising engineered chimeric antigen receptors with at least an additional agent to decrease toxic cytokine release or "cytokine release syndrome” (CRS) or "severe cytokine release syndrome” (sCRS) or "cytokine storm" in the subject.
  • CRS cytokine release syndrome
  • sCRS severe cytokine release syndrome
  • a method of increasing or lengthening the survival of a subject having neoplasia comprising the step of administering to said subject any of the compositions as described herein.
  • a method of preventing neoplasia in a subject comprising the step of administering to said subject any of the compositions as described herein.
  • the neoplasia is selected from the group consisting of blood cancer, B cell leukemia, multiple myeloma, lymphoblastic leukemia (ALL), chronic lymphocytic leukemia, non-Hodgkin's lymphoma, ovarian cancer, or a combination thereof.
  • B cell leukemia multiple myeloma
  • ALL lymphoblastic leukemia
  • chronic lymphocytic leukemia non-Hodgkin's lymphoma
  • ovarian cancer or a combination thereof.
  • the blood cancer is selected from the group consisting of B cell leukemia, multiple myeloma, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia, and non-Hodgkin's lymphoma.
  • ALL acute lymphoblastic leukemia
  • non-Hodgkin's lymphoma non-Hodgkin's lymphoma
  • disclosed herein is a method of treating a cancer or a tumor in a subject, said method comprising the step of administering to said subject any of the compositions as described herein.
  • a method of preventing a cancer or a tumor in a subject said method comprising the step of administering to said subject any of the compositions as described herein.
  • a method of inhibiting a cancer or a tumor in a subject said method comprising the step of administering to said subject any of the compositions as described herein.
  • disclosed herein is a method of reducing a cancer or a tumor in a subject, said method comprising the step of administering to said subject any of the compositions as described herein.
  • a method of ameliorating a cancer or a tumor in a subject said method comprising the step of administering to said subject any of the compositions as described herein.
  • a method of alleviating a cancer or a tumor in a subject said method comprising the step of administering to said subject any of the compositions as described herein.
  • a composition comprising apoptotic cells or an apoptotic supernatant during the immunotherapy.
  • said genetically modified immune cells comprise a T-cell, a naive T-cell, a naive CD4+ T-cell, a naive CD8+ T-cell, a natural killer (NK) cell, a dendritic cell, a cytotoxic T lymphocyte (CTL cell), a regulatory T-cell (Treg), a chimeric antigen receptor (CAR) T-cell, or a genetically modified T-cell receptor (TCR) cell.
  • NK natural killer
  • CTL cell cytotoxic T lymphocyte
  • Reg regulatory T-cell
  • CAR chimeric antigen receptor
  • TCR genetically modified T-cell receptor
  • methods of maintaining or increasing the proliferation rate of the genetically modified immune cells does not reduce or inhibit the efficacy of the immunotherapy.
  • methods of maintaining or increasing the proliferation rate of CAR T-cells does not reduce or inhibit the efficacy of the CAR T-cell cancer therapy.
  • methods of maintaining or increasing the proliferation rate of the genetically modified immune cells, for example CAR T-cells decrease or inhibit cytokine production in the subject.
  • a method of decreasing or inhibiting cytokine production in a subject experiencing cytokine release syndrome or cytokine storm or vulnerable to a cytokine release syndrome or cytokine storm decreases or inhibits cytokine production.
  • the method decreases or inhibits pro-inflammatory cytokine production.
  • the method decreases or inhibits at least one pro-inflammatory cytokine.
  • the method does not reduce the efficacy of the CAR T-cell therapy.
  • the methods provided herein comprise administering a T-cell, NK cell, or CTL cell disclosed herein, in in an amount effective to achieve the desired effect, be it palliation of an existing condition or prevention of recurrence.
  • the amount administered is an amount effective in producing the desired effect.
  • An effective amount can be provided in one or a series of administrations.
  • An effective amount can be provided in a bolus or by continuous perfusion.
  • an "effective amount” may encompass an amount sufficient to effect a beneficial or desired clinical result upon treatment.
  • An effective amount can be administered to a subject in one or more doses.
  • an effective amount is an amount that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of the disease, or otherwise reduce the pathological consequences of the disease.
  • the effective amount is generally determined by the physician on a case-by-case basis and is within the skill of one in the art.
  • factors are typically taken into account when determining an appropriate dosage to achieve an effective amount. These factors include age, sex and weight of the subject, the condition being treated, the severity of the condition and the form and effective concentration of the antigen-binding fragment administered.
  • methods disclosed herein comprise administering a composition comprising a genetically modified cell, and the additional agent or combination thereof, comprised in a single composition.
  • methods comprise administering a composition comprising a CAR T-cell, and the additional agent or combination thereof, comprised in a single composition.
  • methods comprise administering a composition comprising a TCR T-cell, and the additional agent or combination thereof, comprised in a single composition.
  • methods disclosed herein comprise administering a composition comprising a genetically modified cell, and the additional agent or combination thereof, comprised in a at least two compositions.
  • methods comprise administering a composition comprising a CAR T-cell, and the additional agent or combination thereof, comprised in at least two compositions.
  • methods comprise administering a composition comprising a TCR T-cell, and the additional agent or combination thereof, comprised in at least two compositions.
  • T-cells For adoptive immunotherapy using antigen-specific T-cells, for example CAR T-cells, cell doses in the range of 10 6 -10 10 (e.g., 10 9 ) are typically infused. Upon administration of the genetically modified cells into the host and subsequent differentiation, T-cells are induced that are specifically directed against the specific antigen. "Induction" of T-cells may include inactivation of antigen- specific T-cells such as by deletion or anergy. Inactivation is particularly useful to establish or reestablish tolerance such as in autoimmune disorders.
  • the modified cells can be administered by any method known in the art including, but not limited to, intravenous, subcutaneous, intranodal, intratumoral, intrathecal, intrapleural, intraperitoneal and directly to the thymus.
  • the T-cells are not administered intraperitoneally.
  • the T-cells are administered intratumorallly.
  • compositions comprising genetically modified immunoresponsive cells as disclosed herein can be provided systemically or directly to a subject for the treatment of a neoplasia, pathogen infection, or infectious disease.
  • cells disclosed herein are directly injected into an organ of interest (e.g., an organ affected by a neoplasia).
  • compositions comprising genetically modified immunoresponsive cells are provided indirectly to the organ of interest, for example, by administration into the circulatory system (e.g., the tumor vasculature).
  • Expansion and differentiation agents can be provided prior to, during or after administration of the cells to increase production of T-cells, NK cells, or CTL cells in vitro or in vivo.
  • compositions comprising additional agents may be provided prior to, concurrent with, or following administrations of the genetically modified immune cells.
  • genetically modified immune cells for example CAR T-cells are administered prior to an additional agent as disclosed herein.
  • genetically modified immune cells for example CAR T-cells are administered concurrent with an additional agent, as disclosed herein.
  • genetically modified immune cells for example CAR T- cells are administered following administration of an additional agent.
  • methods disclosed herein administer compositions comprising apoptotic cells as disclosed herein. In another embodiment, methods disclosed herein administer compositions comprising apoptotic cell supernatants as disclosed herein.
  • the modified cells can be administered in any physiologically acceptable vehicle, normally intravascularly, although they may also be introduced into bone or other convenient site where the cells may find an appropriate site for regeneration and differentiation (e.g., thymus). Usually, at least lxlO 5 cells will be administered, eventually reaching lxlO 10 or more.
  • Genetically modified immunoresponsive cells disclosed herein may comprise a purified population of cells. Those skilled in the art can readily determine the percentage of genetically modified immunoresponsive cells in a population using various well-known methods, such as fluorescence activated cell sorting (FACS). In some embodiments, ranges of purity in populations comprising genetically modified immunoresponsive cells are about 50 to about 55%, about 55 to about 60%, and about 65 to about 70%.
  • the purity is about 70 to about 75%, about 75 to about 80%, about 80 to about 85%. In further embodiments, the purity is about 85 to about 90%, about 90 to about 95%, and about 95 to about 100%. Dosages can be readily adjusted by those skilled in the art (e.g., a decrease in purity may require an increase in dosage).
  • the cells can be introduced by injection, catheter, or the like. If desired, factors can also be included, including, but not limited to, interleukins, e.g.
  • IL-2 IL-2, IL-3, IL-6, IL-1 1, IL7, IL12, ILIS, IL21, as well as the other interleukins, the colony stimulating factors, such as G-, M- and GM-CSF, interferons, e.g. gamma-interferon and erythropoietin.
  • the colony stimulating factors such as G-, M- and GM-CSF
  • interferons e.g. gamma-interferon and erythropoietin.
  • compositions include pharmaceutical compositions comprising genetically modified immunoresponsive cells or their progenitors and a pharmaceutically acceptable carrier.
  • Administration can be autologous or heterologous.
  • immunoresponsive cells, or progenitors can be obtained from one subject, and administered to the same subject or a different, compatible subject.
  • Peripheral blood derived immunoresponsive cells disclosed herein or their progeny e.g., in vivo, ex vivo or in vitro derived
  • can be administered via localized injection including catheter administration, systemic injection, localized injection, intravenous injection, or parenteral administration.
  • a therapeutic composition as disclosed herein e.g., a pharmaceutical composition containing a genetically modified immunoresponsive cell
  • it will generally be formulated in a unit dosage injectable form (solution, suspension, emulsion).
  • compositions comprising CAR T-cells or other immune cells as disclosed herein are separate from the composition comprising apoptotic cells or an apoptotic supernatant.
  • a method of treating, preventing, inhibiting, reducing the incidence of, ameliorating, or alleviating a malignancy comprising the step of administering a composition comprising chimeric antigen receptor-expressing T-cells (CAR T- cells) and apoptotic cells or an apoptotic cell supernatant.
  • CAR T- cells chimeric antigen receptor-expressing T-cells
  • an anti-tumor immunity response elicited by the genetically modified immune cells may be an active or a passive immune response.
  • the CAR mediated immune response may be part of an adoptive immunotherapy approach in which CAR-modified T-cells induce an immune response specific to the antigen binding moiety in the CAR.
  • immunotherapeutics may encompass the use of immune effector cells and molecules to target and destroy cancer cells.
  • the immune effector may be, for example, an antibody specific for some marker on the surface of a tumor cell.
  • the antibody alone may serve as an effector of therapy or it may recruit other cells to actually effect cell killing.
  • the antibody also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent.
  • the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target.
  • Various effector cells include cytotoxic T cells and NK cells.
  • chimeric antigen receptor-expressing T-cells CAR T-cells
  • these methods may further comprise administering an additional agent in an effort to inhibit or decrease the incidence of CRS or cytokine storm.
  • the cancer is a B-cell malignancy.
  • the B-cell malignancy is leukemia.
  • the B-cell malignancy is acute lymphoblastic leukemia (ALL).
  • ALL acute lymphoblastic leukemia
  • the B-cell malignancy is chronic lymphocytic leukemia.
  • the cancer is leukemia. In one embodiment, the cancer is lymphoma. In one embodiment, the lymphoma is large B-cell lymphoma.
  • the tumor is a solid tumor.
  • a solid tumor is an abnormal mass of tissue lacking cysts or liquid areas.
  • solid tumors are neoplasms (new growth of cells) or lesions (damage of anatomic structures or disturbance of physiological functions) formed by an abnormal growth of body tissue cells other than blood, bone marrow or lymphatic cells.
  • a solid tumor consists of an abnormal mass of cells which may stem from different tissue types such as liver, colon, breast, or lung, and which initially grows in the organ of its cellular origin. However, such cancers may spread to other organs through metastatic tumor growth in advanced stages of the disease.
  • the tumor is a solid tumor.
  • examples of solid tumors are sarcomas, carcinomas, and lymphomas.
  • the solid tumor is an intraperitoneal tumor.
  • the solid tumor comprises an Adrenocortical Tumor (Adenoma and Carcinoma), a Carcinoma, a Colorectal Carcinoma, a Desmoid Tumor, a Desmoplastic Small Round Cell Tumor, an Endocrine Tumor, an Ewing Sarcoma, a Germ Cell Tumor, a Hepatoblastoma a Hepatocellular Carcinoma, a Melanoma, a Neuroblastoma, an Osteosarcoma, a Retinoblastoma, a Rhabdomyosarcoma, a Soft Tissue Sarcoma Other Than Rhabdomyosarcoma, and a Wilms Tumor.
  • Adrenocortical Tumor (Adenoma and Carcinoma)
  • Carcinoma a Colorectal Carcinoma
  • a Desmoid Tumor a Desmoplastic Small Round Cell Tumor
  • an Endocrine Tumor an Ewing Sarcoma
  • the solid tumor is a breast tumor. In another embodiment, the solid tumor is a prostate cancer. In another embodiment, the solid tumor is a colon cancer. In one embodiment, the tumor is a brain tumor. In another embodiment, the tumor is a pancreatic tumor. In another embodiment, the tumor is a colorectal tumor.
  • compositions and methods as disclosed herein have therapeutic and/or prophylactic efficacy against sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endothelio sarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal
  • the tumor is a hematological tumor.
  • hematological tumors are cancer types affecting blood, bone marrow, and lymph nodes. Hematological tumors may derive from either of the two major blood cell lineages: myeloid and lymphoid cell lines.
  • the myeloid cell line normally produces granulocytes, erythrocytes, thrombocytes, macrophages, and masT-cells, whereas the lymphoid cell line produces B, T, NK and plasma cells.
  • Lymphomas e.g.
  • lymphocytic leukemias and myeloma are derived from the lymphoid line, while acute and chronic myelogenous leukemia (AML, CML), myelodysplastic syndromes and myeloproliferative diseases are myeloid in origin.
  • AML, CML acute and chronic myelogenous leukemia
  • myelodysplastic syndromes myeloproliferative diseases are myeloid in origin.
  • compositions and methods as disclosed herein have therapeutic and/or prophylactic efficacy against leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblasts leukemia, acute promyelocyte leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (Hodgkin's disease, non-Hodgkin's disease), Waldenstrom's macroglobulinemia, heavy chain disease.
  • leukemias e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblasts leukemia, acute promyelocyte leukemia, acute myelomonocytic leukemia, acute monocytic le
  • a fragment may encompass at least 5, 10, 13, or 15 amino acids. In other embodiments a fragment is at least 20 contiguous amino acids. Fragments disclosed herein can be generated by methods known to those skilled in the art or may result from normal protein processing (e.g., removal of amino acids from the nascent polypeptide that are not required for biological activity or removal of amino acids by alternative mRNA splicing or alternative protein processing events).
  • antibody and “immunoglobulin” are used interchangeably in the broadest sense and specifically refer to a polyclonal antibody, a monoclonal antibody, or any fragment thereof, which retains the binding activity of the antibody.
  • methods disclosed herein comprise use of a chimeric antibody, a humanized antibody, or a human antibody.
  • polyclonal antibody may encompass a population of different antibodies directed against different determinants (epitopes) of the same antigen.
  • monoclonal antibody may encompass a population of substantially homogenous antibodies, i.e., the individual antibodies comprising the population are identical except for possibly naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are directed against a single antigenic site.
  • the monoclonal antibodies disclosed herein can be made using the hybridoma method first described by Kohler et al, Nature, 256: 495 (1975), or may be made by recombinant DNA methods (e.g. U.S. Pat. No. 4,816,567).
  • a mouse or other appropriate host animal such as a hamster
  • Antibodies to the protein of interest generally are raised in animals by subcutaneous (sc) or intraperitoneal (ip) injections of the desired protein of interest and an adjuvant.
  • the animals are immunized with the protein of interest coupled to Keyhole limpet hemocyanin (KLH, Sigma Aldrich) as a carrier protein.
  • KLH Keyhole limpet hemocyanin
  • the protein of interest used for animal immunization are prepared using methods well- known in the art.
  • the protein of interest may be produced by recombinant methods or by peptide synthesis methods.
  • lymphocytes may be immunized in vitro and then fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • a suitable fusing agent such as polyethylene glycol
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson et al., Anal Biochem., 107: 220 (1980).
  • the antibodies disclosed herein can be produced by using combinatorial libraries to screen for synthetic antibody clones with the desired activity.
  • synthetic antibody clones are selected by screening phage libraries containing phage that display various fragments of antibody variable region (Fv) fused to phage coat protein using methods well known in the art.
  • Fv antibody variable region
  • any fragment thereof which retains the binding activity of the antibody may encompass a portion of an antibody, which may comprise the antigen-binding or variable region thereof, which is capable of binding to the target antigen of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab')2, and Fv fragments.
  • Fab antigen-binding fragments
  • Fc fragment. Pepsin treatment yields an F(ab')2, fragment that has two antigen-combining sites and is still capable of cross- linking antigen.
  • Fv is the minimum antibody fragment which contains a complete antigen-recognition and binding site.
  • polyclonal antibodies and the monoclonal antibodies disclosed herein are prepared using methods well known in the art.
  • a CAR T-cell or related composition in which the CAR is endogenous to the T-cell.
  • endogenous comprises a nucleic acid molecule (e.g., a cDNA, DNA or RNA molecule) or polypeptide that is normally expressed in a cell or tissue.
  • exogenous comprises a nucleic acid molecule or polypeptide that is not endogenously present in the cell, or not present at a level sufficient to achieve the functional effects obtained when artificially over-expressed.
  • exogenous would therefore encompass any recombinant nucleic acid molecule or polypeptide expressed in a cell, such as foreign, heterologous, and over- expressed nucleic acid molecules and polypeptides.
  • CAR T-cells in which the T-cell is autologous to the subject.
  • the CAR T-cells are heterologous to the subject.
  • the CAR T-cells are allogeneic.
  • the CAR T-cells are universal allogeneic CAR T-cells.
  • the T- cells may be autologous, allogeneic, or derived in vitro from engineered progenitor or stem cells.
  • the CAR T-cells and apoptotic cells described herein are both derived from the same source.
  • the CAR T-cells and apoptotic cells described herein are both derived from the subject ( Figure 2B).
  • the CAR T-cells and apoptotic cells described herein are derived from different sources.
  • the CAR T-cells are autologous and the apoptotic cells described herein, are allogeneic ( Figure 3).
  • an apoptotic cell supernatant may be made from cells derived from the same source as the CAR T-cell, which may in one embodiment be autologous cells, or an apoptotic cell supernatant may be made from cells derived from a source different from the source of CAR T-cells.
  • apoptotic cell supernatants may be obtained from different sources.
  • an apoptotic supernatant is obtained from cells undergoing apoptosis.
  • an apoptotic supernatant is obtained from a combination cell cultures wherein apoptotic cells are co-cultured with macrophages and the supernatant is collected.
  • a donor comprises a HLA matched donor. In some embodiments, a donor is an unmatched HLA donor.
  • heterologous may encompass a tissue, cell, nucleic acid molecule or polypeptide that is derived from a different organism.
  • a heterologous protein is a protein that was initially cloned from or derived from a different T-cell type or a different species from the recipient and that is not normally present in a cell or sample obtained from a cell.
  • autologous may encompass a tissue, cell, nucleic acid molecule or polypeptide in which the donor and recipient is the same person.
  • allogeneic may encompass a tissue, cell, nucleic acid molecule or polypeptide that is derived from separate individuals of the same species.
  • allogeneic donor cells are genetically distinct from the recipient.
  • compositions and methods as disclosed herein utilize combination therapy with apoptotic cells or apoptotic supernatants as disclosed herein, and one or more CTLA- 4-blocking agents such as Ipilimumab.
  • CTLA-4 is a potent inhibitor of T-cell activation that helps to maintain self-tolerance.
  • administration of an anti- CTLA-4 blocking agent, which in another embodiment, is an antibody produces a net effect of T- cell activation.
  • compositions and methods as disclosed herein utilize combined therapy comprising apoptotic cells, CAR T-cells, and one or more CTLA-4-blocking agents.
  • a polypeptide of and for use in the methods as disclosed herein comprises at least one conservative amino acid substitution relative to an unmodified amino acid sequence.
  • the polypeptide comprises a non-conservative amino acid substitution.
  • polypeptides having such modifications exhibit increased stability or a longer half-life relative to a polypeptide lacking such an amino acid substitution.
  • methods as disclosed herein may be represented as uses of the compositions as described herein for various therapeutic and prophylactic purposes as described herein, or alternatively, uses of the compositions as described herein in the preparation of a medicament or a therapeutic composition or a composition for various therapeutic and prophylactic purposes as described herein.
  • compositions and methods as disclosed herein comprise the various components or steps.
  • compositions and methods as disclosed herein consist essentially of the various components or steps, where other components or steps may be included.
  • compositions and methods as disclosed herein consist of the various components or steps.
  • the term “comprise” may encompass the inclusion of other components of the composition which affect the efficacy of the composition that may be known in the art.
  • the term “consisting essentially of comprises a composition, which has chimeric antigen receptor-expressing T-cells (CAR T-cells), and apoptotic cells or any apoptotic cell supernatant.
  • CAR T-cells chimeric antigen receptor-expressing T-cells
  • other components may be included that are not involved directly in the utility of the composition.
  • the term "consisting” encompasses a composition having chimeric antigen receptor-expressing T-cells (CAR T-cells), and apoptotic cells or an apoptotic cell supernatant as disclosed herein, in any form or embodiment as described herein.
  • CAR T-cells chimeric antigen receptor-expressing T-cells
  • apoptotic cells or an apoptotic cell supernatant as disclosed herein, in any form or embodiment as described herein.
  • treating comprises therapeutic treatment and “preventing” comprises prophylactic or preventative measures, wherein the object is to prevent or lessen the targeted pathologic condition or disorder as described hereinabove.
  • treating may include directly affecting or curing, suppressing, inhibiting, preventing, reducing the severity of, delaying the onset of, reducing symptoms associated with the disease, disorder or condition, or a combination thereof.
  • "treating,” “ameliorating,” and “alleviating” refer inter alia to delaying progression, expediting remission, inducing remission, augmenting remission, speeding recovery, increasing efficacy of or decreasing resistance to alternative therapeutics, or a combination thereof.
  • preventing refers, inter alia, to delaying the onset of symptoms, preventing relapse to a disease, decreasing the number or frequency of relapse episodes, increasing latency between symptomatic episodes, or a combination thereof.
  • “suppressing” or “inhibiting” refers inter alia to reducing the severity of symptoms, reducing the severity of an acute episode, reducing the number of symptoms, reducing the incidence of disease- related symptoms, reducing the latency of symptoms, ameliorating symptoms, reducing secondary symptoms, reducing secondary infections, prolonging patient survival, or a combination thereof.
  • a composition as disclosed herein is administered once. In another embodiment, the composition is administered twice. In another embodiment, the composition is administered three times. In another embodiment, the composition is administered four times. In another embodiment, the composition is administered at least four times. In another embodiment, the composition is administered more than four times.
  • CAR T-cells as disclosed herein are administered once. In another embodiment, CAR T-cells are administered twice. In another embodiment, CAR T-cells are administered three times. In another embodiment, CAR T-cells are administered four times. In another embodiment, CAR T-cells are administered at least four times. In another embodiment, the composition is administered more than four times.
  • the term "about”, may encompass a deviance of between 0.0001-5% from the indicated number or range of numbers. Further, it may encompass a deviance of between 1 -10% from the indicated number or range of numbers. In addition, it may encompass a deviance of up to 25% from the indicated number or range of numbers.
  • an agent or “at least an agent” may include a plurality of agents, including mixtures thereof.
  • the composition as disclosed herein is a therapeutic composition. In another embodiment, the composition as disclosed herein has therapeutic efficacy.
  • compositions which provides reduced inflammatory cytokine or chemokine release compared to a composition comprising CAR T-cells alone, but with comparable cytotoxicity compared to a composition comprising CAR T-cells alone.
  • the flow chart presented in Figure 1 provides an overview of one embodiment of the steps used during the manufacturing process of a population of early apoptotic cells, wherein anticoagulants are included in the preparation steps. Indicated in the flow chart are the time points at which the anti-coagulants were added during the manufacturing process. As is described in detailed in Example 14 of International Publication No. WO 2014/087408 and United States Application Publication No. US US-2015-0275175-A1, cell populations were prepared wherein anti-coagulants were added at the time of freezing, at the time of incubation, or at the time of freezing and at the time of incubation.
  • Anti-coagulant ACD formula A was supplemented with 10 U/ml heparin at a final concentration of 5% ACD of the total volume and 0.5 U/ml heparin. Methods including anticoagulant consistently produced yields of at least 40% early apoptotic cells, even in the presence of plasma comprising high triglyceride concentrations.
  • Example 11 provide details of preparing another embodiment of apoptotic cell populations that is in the absence of anti-coagulant.
  • Table 3 below shows the comparison of cell populations (batches of cells) prepared with and without anti-coagulant added.
  • the human lymphoma cell line Raji (eCACC, UK, access no. 85011429), the human cervical adenocarcinoma cell line HeLa (ATCC, USA, number: CCL-2) and HeLa-CD19 (ProMab, USA, cat. no. PM-Hela-CD19) were cultured in RPMI 1640 (Gibco, ThermoFisher Scientific, USA, cat. no. 31870-025) supplemented with 10% FBS (Gibco, ThermoFisher Scietific, South America, cat. no. 12657-029), 2 mM GlutaMAX (Gibco, ThermoFisher Scientific, USA, cat. no.
  • HeLa-CD19 medium was further supplemented with 1 ⁇ g/ml puromycin (Sigma-Aldrich, USA, cat. no. P9620), as the selective antibiotics, during standard culturing.
  • PBMCs peripheral blood mononuclear cells
  • target cells (HeLa or HeLa-CD19) were cultured alone or in conjunction with monocytes. After target cells adhered to the plate (6h- overnight), cultures were exposed to y xlO 6 ApoCellsApoCells cells for lh, after which these cells were washed off by 4-5 washes of RPMI. Removal of ApoCells cells was confirmed visually under a light microscope. 10 ng/ml LPS (Sigma-Aldrich, USA, cat. no. L4391) was introduced to the co- culture and incubated for lh. After incubation, LPS was removed by 3-5 washing cycles with RPMI.
  • Viable CD19-CAR T cells or naive T cells were added at the designated E/T ratio(s) and incubated for 4h.
  • plates were centrifuged at 250x g, 2-25°C, 4 min. (Centrifuge 5810 R, Eppendorf, Germany) to sediment cells. 50 ⁇ of supernatant medium from each well was transferred to a fresh flat-bottom 96-well microplate well (Corning, USA, cat. no. 3596) and 50 ⁇ CytoTox 96 Reagent was added to each well. Plates were incubated in the dark at room temperature for 30 min., after which the reaction was terminated by addition of 50 ⁇ Stop Solution per well. Absorbance was read at 492 nm using Infinite F50 (Tecan, Switzerland) and captured using Magellan F50 software. Data analysis and graph generation was performed using Microsoft Excel 2010.
  • Figures 9A through 9H show that there was a significant reduction in the levels of cytokine storm markers IL-10, IL-6, MIP-la, IL-8, TNF-a, ⁇ - ⁇ , MCP-1 , and IL-9 which were induced by LPS in an in vitro model of macrophage activation syndrome.
  • T-cell associated cytokines are not influenced by the CAR T-cell therapy + apoptotic cells, whereas the innate immunity cytokines, for example those released from monocytes, macrophages, and dendritic cells are.
  • EXAMPLE 3 Effect of Apoptotic Cells on Cytokine Storm Without a Negative Effect on The
  • T cells were engineered with a chimeric antigen receptor (CAR) targeting certain ErbB dimers (T4 + CAR-T cells), which are often highly up-regulated in specific solid tumors such as head and neck tumors and ovarian cancers.
  • CAR chimeric antigen receptor
  • T4 + CAR-T cells ErbB dimers
  • T-cells were isolated from PBMC separated from peripheral blood using CD3 micro-beads. Vectors containing the chimeric T4+ receptor were constructed and transducer into the isolated T-cells, resulting in T4+ CAR T-cells.
  • T4+ CAR T-cells were purchased from Creative Biolabs (NY USA) or Promab Biotechnologies (CA USA).
  • Figure 4 presents flow cytometry curves verifying the surface expression of 4 ⁇ chimeric receptor on the T4+ CAR T-cells using an anti- CD124 monoclonal antibody (Wilkie et al., ibid).
  • a PCR procedure was performed and verified the presence of the vector in transduced T cells.
  • SKOV3-luc ovarian adenocarcinoma tissue culture cells were purchased from Cell BioLabs (cat. #AKR-232). SKOV3-luc highly express ErbB receptors and are a target for the T4 + CAR-T cells (van der Stegen et al., 2013, ibid). These cells had been further manipulated to constitutively express the firefly luciferase gene, allowing tracking of cell proliferation in vitro and tumor growth and recession in vivo.
  • PBMCs were isolated using Ficoll (GE healthcare, United Kingdome) from peripheral blood ⁇ buffy coat obtained from healthy, eligible donors. Cells were brought to a concentration of 15xl0 6 cells ⁇ ml in RPMI1640 (Gibco, Thermo Fisher Scientific, MA, USA) and seeded in a 0.9ml drop in the middle of 35mm plates (Corning, NY, USA). Plates were then incubated at 37°C in 5% CO 2 for 1 hour. At the end of incubation, cells were washed three times with PBS (Biological industries, Beit Haemek, Israel) and adhesion was determined using a light microscope. Cells were then incubated with complete media (RPMI1640+ 10% heat inactivated FBS+ 1 % Glutamax+ 1 % PenStrep, all from Gibco).
  • monocyte isolation An alternative method of monocyte isolation was also used wherein human mononuclear cells were isolated from heparinized peripheral blood by density gradient centrifugation. The isolated mononuclear cells then were separated into monocyte, B-cell and T-cell populations by positively selecting monocytes as the CD14+ fraction by magnetic bead separation (Miltenyi Biotec, Auburn, CA, USA), positively selecting B-cells as the CD22+ fraction, and negatively selecting T-cells as the CD14-CD22- fraction. Purity was greater than 95 percent for monocytes.
  • CD14+ monocytes were cultured with apoptotic cells as prepared above at a ratio of 1 :16, for 24h.
  • the number of monocytes was: 0.5 million cells per well in a 12-well plate and the number of apoptotic cells was: 8 million cells per well in a 12-well plate. After incubation for 24 hours the cells were centrifuge (290g, 4 degrees Celsius, 10 minutes). Supernatant was collected and frozen in aliquots at -80 degrees until use. Similar procedures could be performed at different ratios of monocytes :apoptotic cells and/or using other sources of cells, such as macrophages and dendritic cells.
  • lxlO 5 THP-1 cells ml (HTCC USA), or monocytes or macrophages or dendritic cells, will be differentiated with 200 nM (123.4 ng/ml) phorbol myristate acetate (PMA) for 72 hrs and will then be cultured in complete medium without PMA for an additional 24h.
  • PMA phorbol myristate acetate
  • cancer or tumor cells - for example SKOV3-luc cells will be plated in a 24- well plate at 5xl0 5 SKOV3-luc cells/well on the differentiated THP-1 cells.
  • 4xl0 5 -8xl0 5 apoptotic cells (ApoCell) will be added to the culture for 1 -3h to induce an immunotolerant environment.
  • the ratio of cancer cell to ApoCell will be optimized for each cell type.
  • the co-culture will be treated with 10 ng/ml LPS after which lxlO 6 T4 + CAR T cells (or a quantity to be determined by an effector/target (E/T) ratio graph) will be added.
  • the ratios of tumor cells and T4+ CAR T-cells will be varied in order to generate effector/target (E/T) ratio graphs for each tumor or cancer cell type.
  • SKOV3-luc cell lysates were prepared by washing the SKOV3-luc monolayer with PBS to remove any residual serum and adding 70 ⁇ CCLR lysis buffer xl/well (for 24-well plates). Detachment was further enhanced by physical scraping of well bottoms. Following vortexing for 15 seconds, lysates were centrifuged at 12,000g for 2 minutes at 4°C. Supernatants were collected and stored at -80°C.
  • Luciferase Assay System (Promega, cat. #E1501) was used. Calibration of this kit with the luminometer reader (Core Facility, Faculty of Medicine, Ein Kerem, Hebrew University of Jerusalem) was done by using QuantiLum recombinant luciferase (Promega, cat. #E170A). 612 ag - 61.2 ⁇ g (10 ⁇ 20 -10 ⁇ 9 moles) was used to determine detection range and following manufacturer's guidelines. In brief, each rLuciferase quantity in 20 ⁇ volume was placed in a well of black 96-well plates (Nunc). Each quantity was done in triplicate. 100 ⁇ LAR (luciferin substrate from Luciferase Assay System kit) was added to each well and read immediately with a 10 second exposure.
  • LAR luciferin substrate from Luciferase Assay System kit
  • luciferase activity reading For luciferase activity reading, lysates were thawed on ice and 20 ⁇ samples were placed in a black 96-well plate (Nunc). Each sample was read in duplicate. 100 ⁇ LAR was added and luminescence was read for 10 second exposure period every 2.5 minutes for 25 minutes and every 40 seconds for the ensuing 10 minutes.
  • SKOV3-luc growth was followed using luciferase activity as an indicator, to determine target SKOV3-luc cell number in future experiments.
  • 3.8xl0 4 -3.8xl0 5 SKOV3-luc cells/well were plated in 24-well plates (Corning) and luciferase activity was monitored daily for 3 days.
  • 1.9xl0 5 cells/well or higher cell number plated reach confluence and present growth saturation indicated by luciferase activity 2 days after plating ( Figure 5).
  • Note that 3.8xl0 4 -l.lxl0 5 SKOV3-luc cells/well were still in the linear or exponential growth phase three days after plating ( Figure 5, plots orange, turquoise and purple).
  • Negative control (3.8xl0 5 SKOV3-luc cells without LAR substrate) displayed only background-level reading and demonstrates that bioluminescent readings from SKOV3-luc cells result from luciferase activity.
  • T4 + CAR-T cells To corroborate the T4 + CAR-T cell activity, monolayers of SKOV3-luc were exposed to either 1,000,000 (one million) T4 + CAR-T cells or to 1 ,000,000 (one million) non-transduced T cells. After 24h incubation, T4 + CAR-T cells reduced SKOV3-luc proliferation by 30% compared to the non-transduced T cell control ( Figure 6), showing anti-tumor activity of the T4 + CAR-T cells.
  • Apoptotic cells ApoCell
  • Apoptotic cell supernatants ApoSup and ApoMon Sup
  • the SKOV3-luc tumor cells were incubate with Apoptotic Cells for one hour, followed by the addition of T4+ CAR-T cells (500,000, five hundred thousands) or T4+ non-transduced T cells (500,000, five hundred thousands) (ratio of 1 :2 T4 + CAR-T cells to Apoptotic Cells).
  • the tumor cell/Apoptotic cell/T4 + CAR T-cells were then co-cultured for 48h.
  • the control SKOV3-luc tumor cells were co- cultured with T4+ CAR-T cells and Hartman solution (the vehicle of Apoptotic Cells), but without Apoptotic Cells, for 48h.
  • IL-6 is a prototype pro-inflammatory cytokine that is released in cytokine storms (Lee DW et al. (2014) Blood 124(2): 188-195) and is often used as a marker of a cytokine storm response.
  • IL-6 levels measured in the cultured media of SKOV3-luc tumor cells, human monocyte-macrophages, T4+ CAR-T cells, wherein the tumor cells had been previously incubated with apoptotic cells for one hour were dramatically reduced.
  • IL-6 levels measured in the cultured media of SKOV3-luc tumor cells, human monocyte-macrophages, T4+ CAR-T cells, wherein the tumor cells had been previously incubated with apoptotic cell supernatants for one hour were also dramatically reduced. This reduction in concentration of IL-6 is representative of a decrease in the cytokine storm (Figure 8).
  • LPS 10 ng/ml was added to the SKOV3-luc culture conditions outlined above.
  • the addition of LPS is expected to exponentially increase the cytokine storm level.
  • the addition of LPS increased the cytokine storm effect and as a result IL-6 levels increased to approximately 30,000 pg/ml.
  • cytokines known to be expressed in high levels during a cytokine storm showed elevated levels, for example: TNF-a (250-300 pg/ml), IL-10 (200-300 pg/ml), ILl -alpha (40-50 pg/ml) and IL-18 (4-5 pg/ml).
  • TNF-a 250-300 pg/ml
  • IL-10 200-300 pg/ml
  • ILl -alpha 40-50 pg/ml
  • IL-18 4-5 pg/ml
  • the concentration of IL-2 measured in culture supernatants following incubation of SKOV3-luc cells with T4+ CAR T-cells was 1084 pg/ml.
  • concentration of IL-2 increased to 1190 pg/ml.
  • concentration of IL-2R measured in culture supernatants following incubation of SKOV3-luc cells with T4+ CAR T-cells was 3817 pg/ml.
  • SKOC3-luc cells were first incubated with apoptotic cells and then T4+ CAR T-cells the concentration of IL-2R increased to 4580 pg/ml.
  • concentration of 11-2 was 3.2 pg/ml and with the addition of apoptotic cells the concentration was 10.6 pg/ml.
  • concentration of I1-2R was 26.3 pg/ml and with the addition of apoptotic cells the concentration was 24.7 pg/ml.
  • CAR-T cell therapy has been documented to cause cytokine storms in a significant number of patients. These results demonstrate that apoptotic cells and apoptotic cell supernatants surprisingly decreased cytokine storms cytokine markers without affecting CAR-T cell efficacy against tumor cells. Moreover, it appears that apoptotic cells increase cytokine IL-2, which may increase duration of CAR T-cell therapy by maintaining or increasing CAR T-cell proliferation.
  • EXAMPLE 4 Apoptotic Cell Therapy Prevents Cytokine Storms in Mice Administered CAR
  • T4+ CAR T-cells that recognize the ErbB target antigen
  • SKOV3-luc cells apoptotic cells
  • apoptotic supernatants monocytes, macrophages, and the various assays
  • SKOV3-luc tumor cells (1 X 10 6 or 2 X 10 6 ) are inoculated into SCID beige mice or NSGS mice, by either i.p. in PBS or s.c. in 200 ml Matrigel (BD Biosciences). Tumor engraftment is confirmed by bioluminescence imaging (BLI) at about 14-18 days post injection, and mice are sorted into groups with similar signal intensity prior to T-cell administration.
  • BLI bioluminescence imaging
  • mice will receive 30 x 10 6 apoptotic cells either 24 hours prior to administration of T4+ CAR T-cells or concurrent with administration of T4+ CAR T-cells (10-30 x 10 6 T4+ CAR T-cells). Tumor growth will be followed by bioluminescence imaging (BLI) and circulating cytokine levels will be determined by Luminex.
  • BLI bioluminescence imaging
  • Image acquisition parameters were chosen for each image session by imaging mice that received 0.5x10 6 SKOV3-luc cells/mouse, 5 minutes post D-luciferin injection the "auto" option. Capture parameters were set for binning 4, F/stop 1.2 and exposure of 2-4 minutes using the 24x lens. Data analysis and quantification was performed with the Live Image software and graphs were generated using Microsoft's Excel program.
  • mice displayed no clinical symptoms for the initial 4 weeks. However, 28 days post SKOV3-luc injection, the mice that received the high dose (4.5xl0 6 ; purple line) began to lose weighed steadily (Figure 11 A) and the overall appearance of the mice deteriorated, manifested in lethargy, abnormal pacing and general loss of activity. This group was culled at the day 39, and an abdominal autopsy was performed to expose tumor appearance and size (Figure 11B). SKOV3-luc tumors were large, solid, vascularized and displayed a whitish shining complexion. One large tumor predominated on the side of the injection (left) either caudal or rostral in the abdominal cavity.
  • This tumor encompassed approximately 25-75% of the cavity and clearly pressed and disturbed the intestines. Smaller foci were also observed at various locations within the abdominal cavity. Tumors were contained within the abdominal cavity and no other tumors were observed in any other part of the body in any mice. Mice receiving low (0.5x10°) or medium (1x10°) dose of SKOV3-luc cease gaining weight 40 days after SKOV3-luc injection and began to steadily lose weighed. Experiment was terminated 50 days after SKOV3-luc injection.
  • T4+ CAR T-cells Three groups of tumor-free mice as well as mice with tumors are administered (i.p. or directly into the tumor) increasing doses of T4+ CAR T-cells (3xl0 6 , 10 xlO 6 or 30xl0 6 ). At the highest dose, tumor-free mice and mice with tumors demonstrate subdued behavior, piloerection, and reduced mobility within 24 h, accompanied by rapid weight loss followed by death within 48 hrs. At least Human interferon-gamma and mouse IL-6 are detectable in blood samples from the mice given the highest dose of CAR T-cells. Animals that receive a high dose of CAR T-cells directed to a different tumor antigen do not exhibit weight loss or behavioral alterations.
  • mice given the highest dose of CAR T-cells is concomitantly administered 2.10x10 /kg apoptotic cells, which was previously demonstrated to be a safe and effective dose.
  • Mice receiving human CAR T + apoptotic cells have significantly lowered levels of mouse IL-6, lower weight loss, and reduced mortality.
  • CD19+ T4+ CAR T-cells (“CD19+ CAR T-cells")
  • CD19-specific CAR-T cells were purchased from ProMab (Lot # 012916). The T cells were 30% positive for CAR (according to manufacturer's FACS data - Fab staining). Briefly, cells were thawed into AimV + 5% heat-inactivated FBS, centrifuged (300g, 5 minutes, room- temperature),
  • Recombinant HeLa cells expressing CD 19 will be used as a control cell-type that also expresses CD19 on their cell surface.
  • T4+ CAR T-cells will also be engineered with a CAR targeting CD123 epitopes (referred to herein as "CD123+ CAR T-cells").
  • Raji cells Raji cells were purchased from ECACC (Cat. #: 85011429), and routinely cultured in complete medium (RPMI-1640 supplemented with 10% H.I. FBS, 1% Glutamax, 1 %
  • CD19 expressing HeLa cells will be generated in the laboratory and used as a target for CD19+ CAR T-cells.
  • CD123 expressing leukemic cells will be used as targets for
  • CD 123+ CAR T-cells CD 123+ CAR T-cells.
  • primary cancer cells will be utilized as a target for CAR T-cells.
  • HeLa cells expressing CD19 were prepared using methods known in the art. Cells will be cultured as is well known in the art.
  • CD123 is a membrane biomarker and a therapeutic target in hematologic malignancies.
  • CD 123 expressing leukemic cells for example leukemic blasts and leukemic stem cells will be cultured as is known in the art.
  • Apoptotic cells Apoptotic cell supernatants and monocyte isolation, will be prepared as described in Example 1. Early apoptotic cells produced were at least 50% annexin V-positive and less than 5% Pi-positive cells.
  • Macrophages Were generated from CD14positive cells by adherence.
  • Naive T cells were isolated from Buffy coat using magnetic beads (BD), and cryopreserved in 90% human AB serum and 10% DMSO. Thawing and injection was identical to the CAR-T cells.
  • the human lymphoma cell line Raji (eCACC, UK, access no. 85011429), the human cervical adenocarcinoma cell line HeLa (ATCC, USA, number: CCL-2) and HeLa-CD19 (ProMab, USA, cat. no. PM-Hela-CD19) were cultured in RPMI 1640 (Gibco, ThermoFisher Scientific, USA, cat. no. 31870-025) supplemented with 10% FBS (Gibco, ThermoFisher Scietific, South America, cat. no. 12657-029), 2 mM GlutaMAX (Gibco, ThermoFisher Scientific, USA, cat. no.
  • HeLa-CD19 medium was further supplemented with 1 ⁇ puromycin (Sigma-Aldrich, USA, cat. no. P9620), as the selective antibiotics, during standard culturing.
  • PBMCs peripheral blood mononuclear cells
  • CD19-CAR T cells (ProMab, USA, cat. no. FMC63) were delivered either in AIM-V medium or frozen. Cryopreserved CAR T cells for in vitro experiments were thawed on the day of the experiment in a 35-38°C bath and immediately immersed in pre-warmed AIM V medium (Gibco, ThermoFisher Scientific, USA, cat. no. 12055-091) supplemented with 5% FBS (Gibco, South America, cat. no. 12657-029). DMSO was removed by centrifuging the cells (300x g, room temperature, 5 min.) and re-suspending in pre-warmed AIM V medium.
  • CD19-CAR+ cell population Concentration and viability of CD19-CAR+ cell population was determined by anti-FLAG (BioLegend, USA, cat. no. 637310) staining and by Annexin V and PI staining (MEBCYTO Apoptosis kit, MBL, USA, cat. no. 4700) read with FACSCalibur flow cytometer (BD, USA).
  • PBMCs were extracted either from leukapheresis fractions collected from informed consenting eligible donors at Hadassah Medical Center (Ein Kerem Campus, Jerusalem, Israel) using a Cobe SpectraTM apheresis apparatus (Gambro BCT, USA) according to Leaukapheresis Unit's SOP or from buffy coats (Sheba Medical Center, Israel) loaded on a Ficoll density gradient and centrifuged 800x g, 2-8°C, 20 min. T cells were isolated from the positive fraction using MagniSort Human CD3 Positive Selection Kit (eBioscience, USA, cat. no. 8802-6830-74) following manufacturer's guidelines.
  • T cells were cryopreserved in "Complete Medium” (defined above) containing an additional 20% FBS (Gibco, ThermoFisher Scietific, South America, cat. no. 12657-029) and 5% DMSO (CryoSure-DMSO, WAK-Chemie Medical GmbH, Germany, cat. no. WAK-DMSO-70) and thawed on the day of experiment parallel to the CD 19- CAR T cells.
  • FBS Gibco, ThermoFisher Scietific, South America, cat. no. 12657-029
  • DMSO DisoSure-DMSO, WAK-Chemie Medical GmbH, Germany, cat. no. WAK-DMSO-70
  • Lactate dehydrogenase (LDH), a stable cytosolic enzyme, is released by cells undergoing lysis in a correlative manner. Hence, LDH levels in the medium can be used to quantify cytotoxic activity.
  • CytoTox 96 Non-Radioactive Cytotoxicity Assay (Promega, USA, cat. no. G1780) is a colorimetric assay to quantify LDH levels in the medium.
  • a tetrazolium salt substrate iodonitro- tetrazolium violet, INT) is introduced to the medium in excess and LDH converts the substrate into a red formazan product. The amount of red color formed is directly proportional to the number of cells lysed.
  • target cells (HeLa or HeLa-CD19) were cultured alone or in conjunction with monocytes. After target cells adhered to the plate (6h- overnight), cultures were exposed to y xlO 6 ApoCells cells for lh, after which these cells were washed off by 4-5 washes of RPMI. Removal of ApoCells cells was confirmed visually under a light microscope. 10 ng/ml LPS (Sigma-Aldrich, USA, cat. no. L4391) was introduced to the co- culture and incubated for lh. After incubation, LPS was removed by 3-5 washing cycles with RPMI.
  • Viable CD19-CAR T cells or naive T cells were added at the designated E/T ratio(s) and incubated for 4h.
  • plates were centrifuged at 250x g, 2-25°C, 4 min. (Centrifuge 5810 R, Eppendorf, Germany) to sediment cells. 50 ⁇ of supernatant medium from each well was transferred to a fresh flat-bottom 96-well microplate well (Corning, USA, cat. no. 3596) and 50 ⁇ CytoTox 96 Reagent was added to each well. Plates were incubated in the dark at room temperature for 30 min., after which the reaction was terminated by addition of 50 ⁇ Stop Solution per well. Absorbance was read at 492 nm using Infinite F50 (Tecan, Switzerland) and captured using Magellan F50 software. Data analysis and graph generation was performed using Microsoft Excel 2010.
  • HeLa-CD19 (target) and HeLa (control) cells were pre-stained with 5 ⁇ carboxyfluorescein succinimidyl ester (CFSE, Life Technologies, USA, cat. no. CI 157), mixed together, and plated on either fresh plates or on plates populated with isolated primary monocyte. After target cells adhere to the plate (6h-overnight), cultures were exposed to y xlO 6 ApoCells cells for lh. Plates were washed with RPMI 3-5 times and visually verified that suspended ApoCells cells were washed off. 10 ng/ml LPS was introduced to the co-culture and incubated for lh, after which LPS was removed by 3-5 washing cycles with RPMI.
  • CFSE carboxyfluorescein succinimidyl ester
  • Viable CD19-CAR T cells were then added to the co-cultures as indicated by specific E/T ratio(s) and incubated for 4h. After incubation, cells were harvested by adding trypsin-EDTA (Biological Industries, Israel, cat. no. 03-052-1B) and incubating for 4 min. at 37°C. To terminate the enzymatic activity, two- to four-fold volume of "complete medium” was added. Cells were collected, centrifuged at 200x g for 5 min. at room temperature and re-suspended in 100 ⁇ RPMI (Gibco, ThermoFisher Scientific, USA, cat. no. 15140-122). Staining ensued first against anti-CD19 (eBioscience, USA, cat. no.
  • Combination immunotherapy experiments are performed by incubating the Raji cancer cells with apoptotic cells, or apoptotic supernatants, for 1 hour followed by co-culturing with CD 19+ CAR T-cells (+/- monocytes-macrophages) for 48 hours.
  • lxlO 5 THP-1 cells/ml will be differentiated with 200 nM (123.4 ng/ml) phorbol myristate acetate (PMA) for 72 hrs and will then be cultured in complete medium without PMA for an additional 24h.
  • PMA phorbol myristate acetate
  • Raji cancer cells will be plated in a 24-well plate at 5xl0 5 Raji cells/well on the differentiated THP-1 cells.
  • apoptotic cells (ApoCell) will be added to the culture for l-3h to induce an immunotolerant environment.
  • the ratio of cancer cell to ApoCell will be optimized for each cell type.
  • the co-culture will be treated with a pre-determined number of CD19 + CAR-T cells based on the E/T ratio graph.
  • 10 ng/ml LPS will be added to the culture media prior to addition of the CD 19+ CAR T-cells.
  • interferon ⁇ IFN- ⁇
  • the addition of LPS or IFN- ⁇ is expected to exponentially increase the cytokine storm level.
  • Additional cytokine assays examine the level of cytokines IL-10, IL- ⁇ , IL-2, IP-10, IL-4, IL-5, IL-6, IFNa, IL-9, IL-13, IFN- ⁇ , IL-12p70, GM-CSF, TNF-a, MIP-la, ⁇ - ⁇ , IL-17A, IL- 15/IL-15R, or IL-7, or any combination thereof.
  • Raji cells were incubated in the presence of monocytes and LPS, followed by addition of Naive T-cells (Raji + Naive T), CD19+ CAR T-cells (Raji + CAR T), CD19+ CAR T-cells and apoptotic cells (ApoCell) at a ratio of 1 :8 CAR T-cells :ApoCells (Raji + CAR T+ ApoCell 1 :8), CD19+ CAR T-cells and apoptotic cells (ApoCell) at a ratio of 1 :32 CAR T-cells :ApoCells (Raji + CAR T+ ApoCell 1 :32), and CD19+ CAR T-cells and apoptotic cells (ApoCell) at a ratio of 1 :64 CAR T-cells:ApoCells (Raji + CAR T+ ApoCell 1 :64). Concentration measurements were made following GM-
  • Cytokines (mouse or human) may be evaluated by Luminex technology using MAPIX system analyzer (Mereck Millipore)) and MILIPLEX analysis software (Merek Millipore). Mouse IL-6Ra, MIG (CXCL9) and TGF- ⁇ were evaluated by Quantikine ELISA (R&D systems).
  • Bone marrow and liver were evaluated using flow cytometry and immunohistochemistry. Upon sacrifice liver and bone marrow were collected for histopathological analysis. Tissues were fixed in 4% formalin for 48h at room temperature, and then submitted to the animal facility at the Hebrew University for processing. Bones were decalcified prior to processing. Paraffin sections were stained for Hematoxylin and Eosin, and CD19.
  • IFN- ⁇ effect is evaluated both by STAT1 phosphorylation and biological products.
  • CD19 + CAR T-cell activity monolayers of Raji cancer cells are exposed to either 1,000,000 (one million) CD19 + CAR-T cells or to 1,000,000 (one million) non- transduced T cells. After 24h incubation, CD19 + CAR-T cells reduce Raji cancer cell proliferation, showing anti- tumor activity of the CD 19 + CAR-T cells.
  • Apoptotic cells ApoCell
  • Apoptotic cell supernatants ApoSup and ApoMon Sup
  • the Raji Burkett Lymphoma cells are incubate with Apoptotic Cells for one hour, followed by the addition of CD19+ CAR-T cells (500,000, five hundred thousands) or CD19+ non-transduced T cells (500,000, five hundred thousands) (ratio of 1 :2 CD19 + CAR-T cells to Apoptotic Cells).
  • the tumor cell/Apoptotic cell/CD19 + CAR T-cells are then co-cultured for 48h.
  • the control Raji Burkett Lymphoma cells are co-cultured with CD 19+ CAR-T cells and Hartman solution (the vehicle of Apoptotic Cells), but without Apoptotic Cells, for 48h.
  • HeLa cells are specific CD 19 expressing cells, which renders them susceptible to CAR CD19 + T-cell activity.
  • Raji cells which are a non-adherent cell line, HeLa cells are adherent.
  • CD19 + CAR T-cell activity monolayers of HeLa cancer cells were exposed to either 1,000,000 (one million) CD19 + CAR-T cells or to 1,000,000 (one million) non- transduced T cells. After 24h incubation, CD 19 + CAR-T cells reduce HeLa cancer cell proliferation, showing anti-tumor activity of the CD19 + CAR-T cells ( Figure 15 CD19 + + RPMI and CD19 + + CAR T- 19 cells).
  • Apoptotic cells were tested to determine if they interfere with CD 19+ CAR-T cell anti- tumor activity.
  • the HeLa cells were incubated with Apoptotic Cells for one hour, followed by the addition of CD19+ CAR-T cells (500,000, five hundred thousand) or CD19+ non-transduced T cells (Naive T cells; 500,000, five hundred thousand) (ratio of 1 :2 CD19 + CAR-T cells to Apoptotic Cells).
  • the tumor cell/Apoptotic cell/CD19 + CAR T-cells were then co-cultured for 48h.
  • the control HeLa cells were co-cultured with CD 19+ CAR-T cells and RPMI (the vehicle of Apoptotic Cells), but without Apoptotic Cells, for 48h.
  • the CD19 + CAR-T cell:HeLa cell ratio (E T ratio) ranged from 5-20 ( Figure 15).
  • Figure 15 shows that after 48h incubation, CD19+ CAR-T cells anti-tumor activity was superior to incubation with non-transduced T cells (Naive cells) or buffer alone. Similar incubations were performed with apoptotic cells. Surprisingly, CD19 + CAR T-cell anti-tumor activity was comparable with or without exposure to apoptotic cells. Similar experiments are performed using apoptotic cell supernatants. Figure 15 shows the same in vitro cytotoxicity effect of CAR T-CD19 therapy with or without the addition of ApoCells. [0704] No negative effect of the apoptotic cells on CAR-modified T cells against CD19+HeLa cells was observed at comparable E/T ratios in the presence or absence of apoptotic cells.
  • Cytokines IL-8 and IL-13 are measured in the culture media prior to and following addition of CD 19+ CAR T-cells and are showing a concentration consistent with a cytokine storm. Addition of apoptotic cells or apoptotic cell supernatant is showing a reduction of IL-8 and IL-13 concentrations in the media.
  • LPS (10 ng/ml) was added to the Raji cell culture conditions outlined above in the presence of cancer and CAR- 19. The addition of LPS was expected to exponentially increase the cytokine storm level. Exposure to apoptotic cells is dramatically reduced the levels of cytokines.
  • Apoptotic cells were able to down regulate cytokine markers of cytokine storm associated with CAR T-cell clinical procedures. Significantly, the apoptotic cells did not show an effect on the tumor activity of the CAR T-cells. Apoptotic cells decreased pro-inflammatory cytokines that originated from innate immunity and inhibit IFN- ⁇ effect without harming IFN- ⁇ levels and CAR-T cytotoxicity.
  • EXAMPLE 6 Apoptotic Cell Therapy Prevents Cytokine Storms in A Diffuse Cancer in vivo
  • Raji Burkitt lymphoma cells (Sigma-Aldrich cat. # 85011429) were cultured as per the manufacture's guidelines. CD19+ CAR T-cells, cell cultures, apoptotic cells, apoptotic cell supernatants, monocyte isolation, and in vitro measurements are as above for Examples. Early apoptotic cells produced were least 50% annexin V-positive and less than 5% Pi-positive cells.
  • mice 7-8 week old SCID beige mice were purchased from Envigo (formerly known as Harlan). Mice were kept in an SPF free animal facility in compliance with institutional IACUC guidelines. During the course of the experiments the mice were monitored daily, and weighted 3 times a week. Mice showing hind limb paralysis were sacrificed. Upon sacrifice bone marrow and liver were collected for FACS analysis and histological processing, and sera were frozen at -80°C for cytokine profiling. In vivo experiments
  • SCID beige mice C.B-17/IcrHsd-Prkdc-SCID-Lyst-bg, Harlan, Israel
  • AAF The Authority for Animal Facilities
  • AALAC The Hebrew University of Jerusalem
  • AALAC Association for Assessment and Accreditation of Laboratory Animal Care
  • the studies were approved by The Hebrew University Ethics Committee for Animal Experiments, and animal suffering was minimized as possible.
  • mice were examined for clinical indications and weighed twice a week and were sacrificed upon development of hind limb paralysis.
  • Pathological samples of bone and liver were prepared by the Animal Facility Unit of The Hebrew University of Jerusalem and stained against human CD20 (Cell Marque, USA, clone L26, cat. no. 120M-84), to detect Raji cells, and against human CD3 (Cell Signaling Technology, USA, cat. no. 85061), to detect human T cells.
  • CD20 Cell Marque, USA, clone L26, cat. no. 120M-84
  • human CD3 Cell Signaling Technology, USA, cat. no. 85061
  • LPS will be administered to the animal subject prior to addition of the CD 19+ CAR T-cells.
  • interferon- ⁇ IFN- ⁇
  • the addition of LPS or IFN- ⁇ is expected to exponentially increase the cytokine storm level.
  • Cytokine assays examine the level of cytokines including but not limited to IL-10, IL- ⁇ , IL-2, IP-10, IL-4, IL-5, IL-6, IFNa, IL-9, IL-13, IFN- ⁇ , IL-12p70, GM-CSF, TNF-a, MIP-la, MIP- 1 ⁇ , IL-17A, IL-15/IL-15R, or IL-7, or any combination thereof.
  • Cytokines (mouse or human) are evaluated by Luminex technology using MAPIX system analyzer (Mereck Millipore)) and MILIPLEX analysis software (Merek Millipore).
  • Mouse IL-6Ra, MIG (CXCL9) and TGF- ⁇ are evaluated by Quantikine ELISA (R&D systems).
  • Bone marrow and liver are evaluated using flow cytometry and immunohistochemistry. Upon sacrifice liver and bone marrow were collected for histopathological analysis. Tissues were fixed in 4% formalin for 48h at room temperature, and then submitted to the animal facility at the Hebrew University for processing. Bones were decalcified prior to processing. Paraffin sections were stained for Hematoxylin and Eosin, and CD19.
  • IFN- ⁇ effect is evaluated both by STAT1 phosphorylation and biological products.
  • mice Three groups of tumor-free mice as well as mice with tumors are administered (i.p. or directly into the tumor) increasing doses of CD19+ CAR T-cells (3xl0 6 , 10 xlO 6 or 30xl0 6 ).
  • tumor-free mice and mice with tumors demonstrate subdued behavior, piloerection, and reduced mobility within 24 h, accompanied by rapid weight loss followed by death within 48 hrs.
  • Human interferon-gamma, and mouse IL-6 , IL-8, and IL-13 are detectable in blood samples from the mice given the highest dose of CD 19+ CAR T-cells. Animals that receive a high dose of CD 19+ CAR T-cells directed to a different tumor antigen do not exhibit weight loss or behavioral alterations.
  • mice given the highest dose of CD 19+ CAR T-cells is concomitantly administered 2.10xl0 8 /kg apoptotic cells, which was previously demonstrated to be a safe and effective dose.
  • Mice receiving human CD 19+ CAR T + apoptotic cells have significantly lowered levels of at least one mouse pro-inflammatory cytokines, lower weight loss, and reduced mortality.
  • FIG. 18B shows that the expected death of SCID mice injected with CD19 + Raji cells without administration of CD19 + CAR T-cells was 18-21 days. Forty percent (40%) of the mice who received CD19 + CAR T-cells survived to at least day 30 ( Figure 18 blue and yellow lines). The percentage of survivors was independent of the addition of apoptotic cells ( Figure 18). The surviving mice were sacrifice on day 30.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • General Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne des compositions et des méthodes d'utilisation de ces dernières pour inhiber ou réduire l'incidence du syndrome de libération de cytokines ou de la tempête de cytokines chez des sujets traités par une thérapie à base de cellules T CAR, les sujets recevant des compositions comprenant des cellules apoptotiques ou des surnageants de cellules apoptotiques. Dans certains modes de réalisation, les compositions et méthodes d'utilisation de ces dernières décrites dans la description ne diminuent pas l'efficacité du traitement anticancéreux à base de cellules T CAR. L'invention concerne également des compositions et des méthodes d'utilisation de ces dernières pour diminuer ou inhiber la production de cytokines chez un sujet atteint d'un syndrome de libération de cytokines ou d'une tempête de cytokines, comprenant l'administration d'une composition comprenant des cellules apoptotiques ou un surnageant de cellules apoptotiques.
EP17752797.5A 2016-02-18 2017-02-15 Association d'une immunothérapie et d'une thérapie de contrôle des cytokines pour le traitement du cancer Withdrawn EP3416661A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662296622P 2016-02-18 2016-02-18
US201662370741P 2016-08-04 2016-08-04
PCT/IL2017/050196 WO2017141243A1 (fr) 2016-02-18 2017-02-15 Association d'une immunothérapie et d'une thérapie de contrôle des cytokines pour le traitement du cancer

Publications (2)

Publication Number Publication Date
EP3416661A1 true EP3416661A1 (fr) 2018-12-26
EP3416661A4 EP3416661A4 (fr) 2020-03-04

Family

ID=59624865

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17752797.5A Withdrawn EP3416661A4 (fr) 2016-02-18 2017-02-15 Association d'une immunothérapie et d'une thérapie de contrôle des cytokines pour le traitement du cancer

Country Status (9)

Country Link
US (1) US11730761B2 (fr)
EP (1) EP3416661A4 (fr)
JP (2) JP6884155B2 (fr)
KR (1) KR20180110141A (fr)
CN (1) CN109069539A (fr)
AU (1) AU2017219415B2 (fr)
CA (1) CA3014885A1 (fr)
IL (1) IL261008B2 (fr)
WO (1) WO2017141243A1 (fr)

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2974651A1 (fr) 2014-01-24 2015-07-30 Children's Hospital Of Eastern Ontario Research Institute Inc. Polytherapie anticancereuse a base de smc
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
WO2016132366A1 (fr) 2015-02-18 2016-08-25 Enlivex Therapeutics Ltd. Association d'une immunothérapie et d'une thérapie de contrôle des cytokines pour le traitement du cancer
CN114729314A (zh) * 2015-02-18 2022-07-08 伊利威克斯疗法罗德有限公司 用于癌症治疗的组合癌症疗法和细胞因子控制疗法
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US10857181B2 (en) 2015-04-21 2020-12-08 Enlivex Therapeutics Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US11730761B2 (en) 2016-02-18 2023-08-22 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
KR20200014304A (ko) * 2017-06-02 2020-02-10 에프. 호프만-라 로슈 아게 암의 치료를 위한 제ii형 항-cd20 항체 및 항-cd20/항-cd3 이중특이적 항체
EP3612192A4 (fr) * 2017-06-08 2020-05-27 Enlivex Therapeutics Ltd. Cellules apoptotiques thérapeutiques destinées à la thérapie du cancer
US10899831B2 (en) 2017-10-02 2021-01-26 Humanigen, Inc. Method of reducing the level of non-GM-CSF cytokines/chemokines in immunotherapy-related toxicity
US10927168B2 (en) 2017-10-02 2021-02-23 Humanicen, Inc. Method of reducing tumor relapse rate in immunotherapy by administration of lenzilumab
US11130805B2 (en) 2017-10-02 2021-09-28 Humanigen, Inc. Methods of treating CART-T cell therapy-induced neuroinflammation using a GM-CSF antagonist
SG11202003096UA (en) 2017-10-02 2020-05-28 Humanigen Inc Methods of treating immunotherapy-related toxicity using a gm-csf antagonist
CA3082560A1 (fr) * 2017-11-14 2019-05-23 Memorial Sloan-Kettering Cancer Center Cellules immunoreactives secretant il-33 et leurs utilisations
EP3755370A4 (fr) * 2018-02-23 2021-12-01 Cartherics Pty. Ltd. Traitement de maladie des lymphocytes t ciblant la tag-72
WO2020092850A1 (fr) * 2018-10-31 2020-05-07 Humanigen, Inc. Substances et méthodes pour le traitement du cancer
AU2019383076A1 (en) * 2018-11-19 2021-06-10 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use treating sepsis
KR20210124308A (ko) 2019-01-30 2021-10-14 트루바인딩 아이엔씨. 항-gal3 항체 및 이의 용도
CN111544585A (zh) * 2019-02-11 2020-08-18 北京卡替医疗技术有限公司 一种可助推免疫细胞在体内扩增的佐剂
MX2021009786A (es) * 2019-02-14 2021-09-08 Res Institute At Nationwide Children´S Hospital Uso de un agente estimulante para evaluar la potencia de celulas inmunitarias.
CN113189340A (zh) * 2020-01-14 2021-07-30 中国医学科学院基础医学研究所 细胞焦亡通路在细胞治疗中的用途
US20230110998A1 (en) * 2020-02-24 2023-04-13 Yinuoke Medicine Science Technology Company Ltd. Compositions and Methods for Treatment of Cytokine Storm and Cytokine Release Syndrome
CA3185040A1 (fr) * 2020-05-26 2021-12-02 Truebinding, Inc. Methodes de traitement de maladies inflammatoires par blocage de la galectine-3
CN112023048A (zh) * 2020-09-10 2020-12-04 中国医学科学院医学生物学研究所 一种对阿糖胞苷耐药的稳转重组b淋巴细胞白血病细胞系的构建方法及其应用
WO2023150718A2 (fr) * 2022-02-04 2023-08-10 Fred Hutchinson Cancer Center Inhibition par olverembatinib de la régulation à la hausse de cytokines associée au syndrome de libération de cytokines
WO2023156506A1 (fr) * 2022-02-16 2023-08-24 Priothera Sas Méthodes de traitement avec des cellules car en combinaison avec des modulateurs du récepteur s1p
CN116027048A (zh) * 2023-02-06 2023-04-28 山东中鸿特检生物科技有限公司 一种细胞因子风暴综合征诊断试剂盒及其应用

Family Cites Families (174)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4036945A (en) 1976-05-03 1977-07-19 The Massachusetts General Hospital Composition and method for determining the size and location of myocardial infarcts
US4331647A (en) 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5102908A (en) 1985-03-15 1992-04-07 Michael Albeck Method of treating Acquired Immunedeficiency Syndrome (AIDS) using organic tellurium and selenium derivatives
US4962207A (en) 1985-09-30 1990-10-09 Bar-Ilan University Organic derivatives of tellurium and selenium
US4761490A (en) 1985-03-15 1988-08-02 Bar-Ilan University Organic derivatives of tellurium and selenium and their use to stimulate cytokine production
US4752614A (en) 1985-09-30 1988-06-21 Bar-Ilan University Pharmaceutical compositions of tellerium and selenium compounds for the induction of in vivo and in vitro production of cytokines
US4764461A (en) 1985-09-30 1988-08-16 Bar-Ilan University Tellurium and selenium compounds for the induction of in vivo and in vitro production of cytokines
US5093135A (en) 1985-09-30 1992-03-03 Michael Albeck Compounds for the induction of in vivo and in vitro production of cytokines
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
JPS6412935A (en) 1987-07-02 1989-01-17 Mitsubishi Electric Corp Constant-speed travel device for vehicle
US4929739A (en) 1988-03-24 1990-05-29 Bar-Ilan University Complexes of tellurium and selenium derivatives
US5906936A (en) 1988-05-04 1999-05-25 Yeda Research And Development Co. Ltd. Endowing lymphocytes with antibody specificity
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
DE69133476T2 (de) 1990-08-29 2006-01-05 GenPharm International, Inc., Palo Alto Transgene Mäuse fähig zur Produktion heterologer Antikörper
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5213899A (en) 1990-12-17 1993-05-25 General Electric Company Room temperature vulcanizable silicone compositions
US8211422B2 (en) 1992-03-18 2012-07-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric receptor genes and cells transformed therewith
WO1993019163A1 (fr) 1992-03-18 1993-09-30 Yeda Research And Development Co, Ltd. Genes de recepteurs chimeres et cellules transformees au moyen desdits genes
US6479258B1 (en) 1995-12-07 2002-11-12 Diversa Corporation Non-stochastic generation of genetic vaccines
DE19736691A1 (de) 1997-08-22 1999-02-25 Michael Prof Dr Med Giesing Verfahren zur Charakterisierung und Identifizierung disseminierter und metastasierter Krebszellen
US6969609B1 (en) 1998-12-09 2005-11-29 The United States Of America As Represented By The Department Of Health And Human Serivces Recombinant vector expressing multiple costimulatory molecules and uses thereof
US7521197B2 (en) 1998-06-05 2009-04-21 Alexis Biotech Limited Method for producing cytotoxic T-cells
US6962702B2 (en) 1998-06-22 2005-11-08 Immunomedics Inc. Production and use of novel peptide-based agents for use with bi-specific antibodies
US20010033839A1 (en) 1999-10-04 2001-10-25 Emilio Barbera-Guillem Vaccine and immunotherapy for solid nonlymphoid tumor and related immune dysregulation
GB9823897D0 (en) 1998-11-02 1998-12-30 Imp College Innovations Ltd Immunotherapeutic methods and molecules
IL127142A0 (en) 1998-11-19 1999-09-22 Yeda Res & Dev Immune cells having predefined biological specificity
US7829064B2 (en) 1999-05-10 2010-11-09 Immunomedics, Inc. Anti-CD74 immunoconjugates and methods
US7550143B2 (en) 2005-04-06 2009-06-23 Ibc Pharmaceuticals, Inc. Methods for generating stably linked complexes composed of homodimers, homotetramers or dimers of dimers and uses
US8383081B2 (en) 1999-05-10 2013-02-26 Immunomedics, Inc. Anti-CD74 immunoconjugates and methods of use
US8119101B2 (en) 1999-05-10 2012-02-21 The Ohio State University Anti-CD74 immunoconjugates and methods of use
US7534866B2 (en) 2005-10-19 2009-05-19 Ibc Pharmaceuticals, Inc. Methods and compositions for generating bioactive assemblies of increased complexity and uses
US7842458B2 (en) 1999-08-12 2010-11-30 Agensys, Inc. Nucleic acids and corresponding proteins entitled 58P1D12 useful in treatment and detection of cancer
US20040214783A1 (en) 2002-05-08 2004-10-28 Terman David S. Compositions and methods for treatment of neoplastic disease
US6436703B1 (en) 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
US6489311B1 (en) 2000-05-02 2002-12-03 Charlotte-Mecklenburg Hospital Authoirty Method for the prevention of apoptosis
CA2309424A1 (fr) 2000-05-25 2001-11-25 Vasogen Ireland Limited Entites apoptotiques pour le traitement de troubles neurodegeneratifs et autres troubles neurologiques
CA2309518A1 (fr) 2000-05-25 2001-11-25 Vasogen Ireland Limited Entites apoptotiques pour le traitement de troubles ou les lymphocytes t jouent un role mediateur et les troubles dits inflammatoires
WO2001097844A1 (fr) 2000-06-22 2001-12-27 Idec Pharmaceuticals Corporation Proteine de fusion bispecifique et son procede d'utilisation permettant d'ameliorer l'action tueuse de cellules effectives sur des cellules cibles
AUPR011700A0 (en) 2000-09-14 2000-10-05 Austin Research Institute, The Composition comprising immunogenic virus sized particles (VSP)
US20030036505A1 (en) 2000-09-25 2003-02-20 Human Genome Sciences, Inc. Signal transduction pathway component polynucleotides, polypeptides, antibodies and methods based thereon
US7491534B2 (en) 2000-12-22 2009-02-17 Kirin Holdings Kabushiki Kaisha Methods for altering cell fate to generate T-cells specific for an antigen of interest
US20050202098A1 (en) 2001-01-31 2005-09-15 Tolaren Disease therapy using dying or dead cells
US7927597B2 (en) 2001-04-10 2011-04-19 Agensys, Inc. Methods to inhibit cell growth
US7811575B2 (en) 2001-04-10 2010-10-12 Agensys, Inc. Nucleic acids and corresponding proteins entitled 158P3D2 useful in treatment and detection of cancer
US20020193569A1 (en) 2001-06-04 2002-12-19 Idec Pharmaceuticals Corporation Bispecific fusion protein and method of use for enhancing effector cell killing of target cells
GB0123379D0 (en) 2001-09-28 2001-11-21 Lorantis Ltd Modulators
BR0213303A (pt) 2001-10-15 2005-06-07 Immunomedics Inc Proteìnas de ligação de alvejamento direto
US8877901B2 (en) 2002-12-13 2014-11-04 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US7591994B2 (en) 2002-12-13 2009-09-22 Immunomedics, Inc. Camptothecin-binding moiety conjugates
KR20040088572A (ko) 2002-03-01 2004-10-16 이뮤노메딕스, 인코오포레이티드 제거율 증강을 위한 양특이성 항체 점 돌연변이들
DE60333732D1 (de) 2002-03-01 2010-09-23 Immunomedics Inc Internalisierung von anti cd74 monoklonalen antikörpern und deren verwendungen
WO2003075952A1 (fr) 2002-03-05 2003-09-18 Board Of Regents, The University Of Texas System Procedes d'amelioration de l'induction de la reponse immunitaire impliquant mda-7
US7993626B2 (en) 2007-01-11 2011-08-09 Immunomedics, Inc. Methods and compositions for F-18 labeling of proteins, peptides and other molecules
WO2004016733A2 (fr) 2002-08-16 2004-02-26 Agensys, Inc. Acides nucleiques et proteines correspondantes connues sous 251p5g2 que l'on utilise dans le traitement et la detection de cancers
AU2003282306A1 (en) 2002-10-29 2004-05-25 Engene, Inc. Compositions for cancer treatment
US7534427B2 (en) 2002-12-31 2009-05-19 Immunomedics, Inc. Immunotherapy of B cell malignancies and autoimmune diseases using unconjugated antibodies and conjugated antibodies and antibody combinations and fusion proteins
CN1761757A (zh) 2003-01-07 2006-04-19 香港大学 用血管抑制素协同加强腺伴随病毒介导的b7.1免疫接种以根除扩散的肝转移性肿瘤
EP1590437A4 (fr) 2003-01-24 2008-06-18 Agensys Inc Acides nucleiques et proteines correspondantes dites 254p1d6b utiles pour la detection et le traitement du cancer
US20040202666A1 (en) 2003-01-24 2004-10-14 Immunomedics, Inc. Anti-cancer anthracycline drug-antibody conjugates
US20050008618A1 (en) 2003-02-27 2005-01-13 Howard Kaufman Composition for delivering an agent to a target cell and uses thereof
WO2004093808A2 (fr) 2003-04-22 2004-11-04 Maxygen, Inc. Nouveaux antigenes associes a une tumeur
AU2004235755A1 (en) 2003-04-30 2004-11-18 Agensys, Inc. Nucleic acids and corresponding proteins entitled 109P1D4 useful in treatment and detection of cancer
JP2007525196A (ja) 2003-05-30 2007-09-06 アジェンシス, インコーポレイテッド 前立腺幹細胞抗原(psca)変異体及びその部分配列
US20070248584A1 (en) 2003-06-10 2007-10-25 The University Of Melbourne Immunomodulating Compositions, Uses Therefore and Processes for Their Production
CA2534898A1 (fr) 2003-08-08 2005-02-17 Immunomedics, Inc. Anticorps bispecifiques pour induire l'apoptose de cellules tumorales et malades
WO2005019429A2 (fr) 2003-08-22 2005-03-03 Potentia Pharmaceuticals, Inc. Compositions et procedes permettant d'augmenter la phagocytose ou l'activite des phagocytes
WO2005117846A2 (fr) 2004-05-27 2005-12-15 Vertex Pharmaceuticals Incorporated Traitement de maladies a l'aide d'inhibiteurs de l'ice
WO2005049852A2 (fr) 2003-11-17 2005-06-02 University Of Florida Procedes et compositions destines a induire l'apoptose
WO2005052119A2 (fr) 2003-11-19 2005-06-09 Beth Israel Deaconess Medical Center Adjuvants de la reponse immunitaire
CN1934072A (zh) 2004-01-30 2007-03-21 派普林生物脂股份有限公司 治疗和载体分子
WO2007046893A2 (fr) 2005-10-19 2007-04-26 Ibc Pharmaceuticals, Inc. Procedes et compositions permettant de produire des ensembles bioactifs de complexite augmentee et utilisations
US7674456B2 (en) 2004-06-14 2010-03-09 Charles Wiseman Breast cancer cell lines and uses thereof
GB0414055D0 (en) 2004-06-23 2004-07-28 Univ Edinburgh Specific binding members and uses thereof
WO2006004620A2 (fr) 2004-06-29 2006-01-12 Terman David S Superantigenes a base d'amas de genes de l'enterotoxine pour le traitement de maladie maligne
JP2008509700A (ja) 2004-08-16 2008-04-03 アジェンシス, インコーポレイテッド 癌の処置および検出において有用な58p1d12と称される核酸および対応するタンパク質
DE602005008622D1 (de) 2004-09-17 2008-09-11 Biomas Ltd Neue tellurium-verbindungen und ihre verwendung als immunmodulatoren
WO2006055004A1 (fr) 2004-11-19 2006-05-26 Agensys, Inc. Acides nucléiques et protéines correspondantes désignés par 158p3d2, et pouvant être employés dans le traitement et la détection de cancers
WO2006063150A2 (fr) 2004-12-08 2006-06-15 Immunomedics, Inc. Procedes et compositions pour l'immunotherapie et la detection de maladies inflammatoires et de dereglement immunitaire, de maladies infectieuses, d'angiogeneses pathologiques et de cancers
CA2594356C (fr) 2005-01-05 2018-07-17 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H. Domaines immunoglobuline synthetiques a proprietes de liaison elabores dans des regions de la molecule differentes des regions de determination de complementarite
DE102005013846A1 (de) 2005-03-24 2006-10-05 Ganymed Pharmaceuticals Ag Identifizierung von Oberflächen-assoziierten Antigenen für die Tumordiagnose und -therapie
JP5011277B2 (ja) 2005-04-06 2012-08-29 アイビーシー・ファーマシューティカルズ・インコーポレーテッド ホモダイマー、ホモテトラマーまたはダイマーのダイマーのからなる安定に連結された複合体を発生させるための方法および使用
EP1896570A2 (fr) 2005-05-10 2008-03-12 Avaris AB Nouvelles compositions et leurs utilisations
WO2006135454A1 (fr) 2005-06-08 2006-12-21 Saint Vincent Medical Center, A California Corporation Nouvelles lignees de cellules cancereuses et utilisations correspondantes
MX2009000285A (es) 2006-06-30 2009-06-08 Schering Corp Piperidinas sustituidas que incrementan la actividad de p53 y usos de las mismas.
US20080081791A1 (en) 2006-07-06 2008-04-03 Weida Huang Methods of using combinations of siRNAs for treating a disease or a disorder, and for enhancing siRNA efficacy in RNAi
GB0622399D0 (en) 2006-11-10 2006-12-20 Avaris Ab Novel compositions and uses thereof
LT2126054T (lt) 2007-01-31 2016-10-10 Yeda Research And Development Company Limited Peradresuotos, genetiškai modifikuotos t reguliavimo ląstelės ir jų naudojimas autoimuninės ir uždegiminės ligos slopinimui
WO2008137901A2 (fr) 2007-05-06 2008-11-13 Sloan Kettering Institute For Cancer Research Procédés de traitement et de prévention du syndrome gi et de la réaction de greffe contre l'hôte
CA2695385A1 (fr) 2007-07-31 2009-02-05 Atul Bedi Conjugue d'acide nucleique de polypeptide destine a l'immunoprophylaxie ou a l'immunotherapie des troubles neoplasiques ou infectieux
US9585957B2 (en) 2007-09-07 2017-03-07 The Johns Hopkins University Adenosine receptor agonists and antagonists to modulate T cell responses
US10736848B2 (en) 2007-10-12 2020-08-11 Massachusetts Institute Of Technology Vaccine nanotechnology
US20090192114A1 (en) 2007-12-21 2009-07-30 Dmitriy Ovcharenko miR-10 Regulated Genes and Pathways as Targets for Therapeutic Intervention
WO2009083009A2 (fr) 2008-01-03 2009-07-09 Genmab A/S Anticorps monoclonaux anti-cd32b
US9272029B2 (en) 2009-03-26 2016-03-01 Ibc Pharmaceuticals, Inc. Interferon lambada-antibody complexes
WO2010024852A1 (fr) 2008-08-28 2010-03-04 Nestec S.A. Profils d’expression génique associés avec un phénotype maigre et leurs utilisations
CN102753193A (zh) 2008-10-31 2012-10-24 比奥根艾迪克Ma公司 Light靶向分子及其用途
AU2009325878B2 (en) 2008-12-08 2014-01-16 Compugen Ltd. TMEM154 polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
UA109633C2 (uk) 2008-12-09 2015-09-25 Антитіло людини проти тканинного фактора
EP2201954A1 (fr) 2008-12-18 2010-06-30 Aposcience AG Préparation pharmaceutique
HUE060624T2 (hu) 2009-02-13 2023-04-28 Immunomedics Inc Sejten belüli hasítható kötést tartalmazó immunkonjugátumok
US8506954B2 (en) 2009-12-01 2013-08-13 The Board Of Trustees Of The Leland Stanford Junior University Tumor vaccination in combination with hematopoietic cell transplantation for cancer therapy
EP2524059A4 (fr) 2010-01-13 2013-11-20 Caris Life Sciences Luxembourg Holdings Détection d'affections gastro-intestinales
AU2011223789A1 (en) 2010-03-01 2012-09-20 Caris Life Sciences Switzerland Holdings Gmbh Biomarkers for theranostics
IL300733A (en) 2010-03-05 2023-04-01 Univ Johns Hopkins Compositions and methods for antibodies and fusion proteins targeting immune modulation
CN103003307B (zh) 2010-03-10 2017-08-11 根马布股份公司 抗c‑MEt的单克隆抗体
GB201004551D0 (en) 2010-03-19 2010-05-05 Immatics Biotechnologies Gmbh NOvel immunotherapy against several tumors including gastrointestinal and gastric cancer
MX2012011688A (es) 2010-04-09 2012-11-23 Amgen Inc Proteinas btnl9, acidos nucleicos y anticuerpos y usos de los mismos.
WO2011139629A2 (fr) 2010-04-26 2011-11-10 Biogen Idec Ma Inc. Molécules ciblant light et ses utilisations
EP2566954B2 (fr) 2010-05-04 2022-11-02 Yeda Research and Development Co. Ltd. Immunothérapie à base de cellules allogéniques redirigées
EP2387999A1 (fr) 2010-05-21 2011-11-23 CureVac GmbH Solution contenant de l'histidine pour la transfection et/ou l'injection d'acides nucléiques et utilisations associées
JP6320753B2 (ja) 2010-05-27 2018-05-09 ゲンマブ エー/エス Her2に対するモノクローナル抗体
WO2012024543A1 (fr) 2010-08-18 2012-02-23 Caris Life Sciences Luxembourg Holdings Biomarqueurs circulants pour une maladie
EP2614143B1 (fr) * 2010-09-08 2018-11-07 Baylor College Of Medicine Immunothérapie des cancers bronchique non à petites cellules utilisant des lymphocytes t génétiquement modifiés, spécifiques de gd2
CN103282048B (zh) 2010-10-01 2017-05-17 宾夕法尼亚大学理事会 李斯特菌疫苗载体用于在寄生虫感染的个体中扭转免疫无应答的用途
WO2012088302A2 (fr) 2010-12-22 2012-06-28 Abbott Laboratories Protéines de liaison à une demi-immunoglobuline et leurs utilisations
US20120196762A1 (en) 2011-01-27 2012-08-02 Paradis Norman A Method and apparatus for discovery, development and clinical application of multiplex assays based on patterns of cellular response
US20150025812A1 (en) 2011-01-27 2015-01-22 Norman A. Paradis Method and apparatus for discovery, development and clinical application of multiplex assays based on patterns of cellular response
CA2826186C (fr) 2011-02-01 2020-08-04 Genmab A/S Anticorps humains et conjugues anticorps-medicament contre cd74
US20140141986A1 (en) 2011-02-22 2014-05-22 David Spetzler Circulating biomarkers
KR20140023903A (ko) 2011-02-24 2014-02-27 온코타이레온, 인코포레이티드 항원보강제를 갖는 muc1 기초 당지질펩티드 백신
AU2012240135B2 (en) 2011-04-08 2016-09-08 Baylor College Of Medicine Reversing the effects of the tumor microenvironment using chimeric cytokine receptors
BR112013032232A2 (pt) 2011-06-16 2016-09-20 Caris Life Sciences Switzerland Holdings S A R L método de caracterização de câncer através do uso de biomarcador de ácido nucleico
KR20140067001A (ko) 2011-08-08 2014-06-03 카리스 라이프 사이언스 룩셈부르크 홀딩스, 에스.에이.알.엘. 생물지표 조성물 및 방법
SG10201607663YA (en) 2011-08-17 2016-11-29 Globeimmune Inc Yeast-muc1 immunotherapeutic compositions and uses thereof
CA2856895C (fr) 2011-11-28 2021-10-26 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associees
EP2802599B1 (fr) 2012-01-09 2019-09-04 Yeda Research and Development Co. Ltd. Peptides anti-inflammatoires et leur utilisation
WO2013112801A1 (fr) 2012-01-26 2013-08-01 Ibc Pharmaceuticals, Inc. Ciblage d'interféron-lambda par des anticorps favorisant puissamment les activités antivirales et antitumorales.
CN107496932A (zh) 2012-02-27 2017-12-22 阿穆尼克斯运营公司 Xten缀合组合物和制造其的方法
EP3173788A3 (fr) 2012-03-14 2017-07-12 Marx, Stephen Moyens et procédés pour le diagnostic et la thérapeutique de maladies
US10130714B2 (en) 2012-04-14 2018-11-20 Academia Sinica Enhanced anti-influenza agents conjugated with anti-inflammatory activity
US20130280220A1 (en) 2012-04-20 2013-10-24 Nabil Ahmed Chimeric antigen receptor for bispecific activation and targeting of t lymphocytes
AU2013278843A1 (en) 2012-06-21 2014-03-27 Compugen Ltd. LSR antibodies, and uses thereof for treatment of cancer
DK4019041T3 (da) * 2012-07-13 2023-04-03 Univ Pennsylvania Toksicitetshåndtering for cars antitumoraktivitet
US9682143B2 (en) 2012-08-14 2017-06-20 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
AU2013302696B9 (en) 2012-08-14 2018-08-09 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
WO2014055657A1 (fr) 2012-10-05 2014-04-10 The Trustees Of The University Of Pennsylvania Utilisation d'une approche trans-signalisation dans des récepteurs d'antigènes chimériques
AU2013340414B2 (en) 2012-10-23 2019-05-02 Caris Science, Inc. Aptamers and uses thereof
US9717731B2 (en) 2012-11-02 2017-08-01 Pharmacyclics Llc TEC family kinase inhibitor adjuvant therapy
KR20150086294A (ko) 2012-11-05 2015-07-27 프로나이 테라퓨틱스, 인코포레이티드 올리고뉴클레오티드 암 치료제의 복용 및 투여
US10131682B2 (en) 2012-11-24 2018-11-20 Hangzhou Dac Biotech Co., Ltd. Hydrophilic linkers and their uses for conjugation of drugs to a cell binding molecules
US20150301058A1 (en) 2012-11-26 2015-10-22 Caris Science, Inc. Biomarker compositions and methods
CA2893962C (fr) * 2012-12-06 2021-04-06 Enlivex Therapeutics Ltd Preparations de cellules apoptotiques therapeutiques, methode de production et utilisations associees
WO2014106666A1 (fr) 2013-01-07 2014-07-10 Institut National De La Sante Et De La Recherche Medicale (Inserm) Traitement thérapeutique consistant à utiliser une préparation pharmaceutique tolérogénique
EP2953524B1 (fr) 2013-02-06 2018-08-01 Freenome Holdings Inc. Systèmes et procédés pour la détection de maladie précoce et la surveillance de maladie en temps réel
CA2904369A1 (fr) * 2013-03-07 2014-09-12 Baylor College Of Medicine Ciblage de cd138 dans le cancer
JP2016513458A (ja) 2013-03-10 2016-05-16 ベイラー カレッジ オブ メディスンBaylor College Of Medicine 化学療法耐性免疫細胞
EP2970985A1 (fr) 2013-03-14 2016-01-20 Fred Hutchinson Cancer Research Center Compositions et méthodes de modification de cellules à des fins thérapeutiques
US9657105B2 (en) 2013-03-15 2017-05-23 City Of Hope CD123-specific chimeric antigen receptor redirected T cells and methods of their use
JP6480910B6 (ja) 2013-03-15 2019-04-03 ザ・チルドレンズ・ホスピタル・オブ・フィラデルフィアThe Children’S Hospital Of Philadelphia 無血清懸濁培養物系で組み換えレンチウイルスベクターを産生するための拡張可能な生産プロセス
JP2016512833A (ja) 2013-03-15 2016-05-09 ゲームス・ファーマ・インコーポレイテッド ウイルス感染症の治療薬としてのベラプロスト異性体
CA2899577C (fr) 2013-04-03 2023-10-17 Ibc Pharmaceuticals, Inc. Polytherapie pour induire une reponse immunitaire a une maladie
US10584158B2 (en) 2013-04-17 2020-03-10 Baylor College Of Medicine Immunosuppressive TGF-β signal converter
US20160090638A1 (en) 2013-05-17 2016-03-31 National Health Research Institutes Methods of prognostically classifying and treating glandular cancers
WO2014193999A2 (fr) 2013-05-28 2014-12-04 Caris Science, Inc. Méthodes et compositions d'identification de biomarqueurs
ES2791598T3 (es) 2013-06-05 2020-11-05 Bellicum Pharmaceuticals Inc Métodos para inducir apoptosis parcial utilizando polipéptidos de caspasa
WO2014201021A2 (fr) 2013-06-10 2014-12-18 Dana-Farber Cancer Institute, Inc. Procédés et compositions pour réduire l'immunodépression par des cellules tumorales
JP6585041B2 (ja) 2013-07-18 2019-10-02 ベイラー カレッジ オブ メディスンBaylor College Of Medicine 免疫細胞の能力を増強する方法
MX2016007748A (es) 2013-12-13 2017-07-28 Angiogenex Inc Composiciones y métodos para tratar, prevenir y diagnosticar cáncer y otros trastornos proliferativos.
CA2946511C (fr) * 2014-04-21 2023-08-22 The Childen's Hospital Of Philadelphia Compositions et methodes pour le traitement de troubles lies aux cytokines
WO2016132366A1 (fr) 2015-02-18 2016-08-25 Enlivex Therapeutics Ltd. Association d'une immunothérapie et d'une thérapie de contrôle des cytokines pour le traitement du cancer
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US10857181B2 (en) * 2015-04-21 2020-12-08 Enlivex Therapeutics Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US11730761B2 (en) 2016-02-18 2023-08-22 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
EP3612192A4 (fr) 2017-06-08 2020-05-27 Enlivex Therapeutics Ltd. Cellules apoptotiques thérapeutiques destinées à la thérapie du cancer

Also Published As

Publication number Publication date
JP7223055B2 (ja) 2023-02-15
JP2019505552A (ja) 2019-02-28
IL261008A (en) 2018-10-31
IL261008B2 (en) 2023-07-01
US11730761B2 (en) 2023-08-22
IL261008B1 (en) 2023-03-01
EP3416661A4 (fr) 2020-03-04
KR20180110141A (ko) 2018-10-08
AU2017219415B2 (en) 2023-08-10
JP6884155B2 (ja) 2021-06-09
WO2017141243A1 (fr) 2017-08-24
JP2021119197A (ja) 2021-08-12
CA3014885A1 (fr) 2017-08-24
US20190175649A1 (en) 2019-06-13
CN109069539A (zh) 2018-12-21
AU2017219415A1 (en) 2018-09-06

Similar Documents

Publication Publication Date Title
AU2017219415B2 (en) Combination immune therapy and cytokine control therapy for cancer treatment
US20230084027A1 (en) Combination immune therapy and cytokine control therapy for cancer treatment
US11717539B2 (en) Combination immune therapy and cytokine control therapy for cancer treatment
AU2020350221A1 (en) Combination cancer therapy and cytokine control therapy for cancer treatment
CN114729314A (zh) 用于癌症治疗的组合癌症疗法和细胞因子控制疗法

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180914

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 16/28 20060101ALI20190905BHEP

Ipc: A61K 39/00 20060101ALI20190905BHEP

Ipc: A61P 35/00 20060101ALI20190905BHEP

Ipc: C12N 5/0783 20100101ALI20190905BHEP

Ipc: A61K 35/17 20150101AFI20190905BHEP

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 16/28 20060101ALI20200121BHEP

Ipc: A61K 35/17 20150101AFI20200121BHEP

Ipc: A61K 39/00 20060101ALI20200121BHEP

Ipc: C12N 5/0783 20100101ALI20200121BHEP

Ipc: A61P 35/00 20060101ALI20200121BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20200205

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ENLIVEX THERAPEUTICS LTD.

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ENLIVEX THERAPEUTICS RDO LTD

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20221206

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230522

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20240123